Vol. 28 (2023)



No. 01  DOI: 10.1186/s11658-022-00409-6 Volume 28 (2023) - 28:01
Title THREE-DIMENSIONAL GENOME LANDSCAPE COMPREHENSIVELY REVEALS PATTERNS OF SPATIAL GENE REGULATION IN PAPILLARY AND ANAPLASTIC THYROID CANCERS: A STUDY USING REPRESENTATIVE CELL LINES FOR EACH CANCER TYPE
Authors Linlin Zhang1†, Miaomiao Xu1†, Wanchun Zhang2†, Chuanying Zhu3, Zhilei Cui4, Hongliang Fu1, Yufei Ma1, Shuo Huang1, Jian Cui5*, Sheng Liang1*, Lei Huang6,7* and Hui Wang1*
Abstract Background: Spatial chromatin structure is intricately linked with somatic aberrations, and somatic mutations of various cancer-related genes, termed co-mutations (CoMuts), occur in certain patterns during cancer initiation and progression. The functional mechanisms underlying these genetic events remain largely unclear in thyroid cancer (TC). With discrepant diferentiation, papillary thyroid cancer (PTC) and anaplastic thyroid cancer (ATC) difer greatly in characteristics and prognosis. We aimed to reveal the spatial gene alterations and regulations between the two TC subtypes.
Methods: We systematically investigated and compared the spatial co-mutations between ATC (8305C), PTC (BCPAP and TPC-1), and normal thyroid cells (Nthy-ori-3–1). We constructed a framework integrating whole-genome sequencing (WGS), high-throughput chromosome conformation capture (Hi-C), and transcriptome sequencing, to systematically detect the associations between the somatic co-mutations of cancer-related genes, structural variations (SVs), copy number variations (CNVs), and high-order chromatin conformation.
Results: Spatial co-mutation hotspots were enriched around topologically associating domains (TADs) in TC. A common set of 227 boundaries were identifed in both ATC and PTC, with signifcant overlaps between them. The spatial proximities of the co-mutated gene pairs in the two TC types were signifcantly greater than in the gene-level and overall backgrounds, and ATC cells had higher TAD contact frequency with CoMuts>10 compared with PTC cells. Compared with normal thyroid cells, in ATC the number of the created novel three-dimensional chromatin structural domains increased by 10%, and the number of shifted TADs decreased by 7%. We found fve TAD blocks with CoMut genes/events specifc to ATC with certain mutations in genes including MAST-NSUN4, AM129B/TRUB2, COL5A1/PPP1R26, PPP1R26/GPSM1/CCDC183, and PRAC2/DLX4. For the majority of ATC and PTC cells, the HOXA10 and HIF2α signals close to the transcription start sites of CoMut genes within TADs were signifcantly stronger than those at the background. CNV breakpoints signifcantly overlapped with TAD boundaries in both TC subtypes. ATCs had more CNV losses overlapping with TAD boundaries, and noncoding SVs involved in intrachromosomal SVs, amplifed inversions, and tandem duplication difered between ATC and PTC. TADs with short range were more abundant in ATC than PTC. More switches of A/B compartment types existed in ATC cells compared with PTC. Gene expression was signifcantly synchronized, and orchestrated by complex epigenetics and regulatory elements.
Conclusion: Chromatin interactions and gene alterations and regulations are largely heterogeneous in TC. CNVs and complex SVs may function in the TC genome by interplaying with TADs, and are largely diferent between ATC and PTC. Complexity of TC genomes, which are highly organized by 3D genome-wide interactions mediating mutational and structural variations and gene activation, may have been largely underappreciated. Our comprehensive analysis may provide key evidence and targets for more customized diagnosis and treatment of TC.
Keywords Papillary thyroid cancer, Anaplastic thyroid cancer, Genomic heterogeneity, Topologically associating domains (TADs), High-throughput chromosome conformation capture (Hi-C), A/B compartment switches, Somatic hotspot mutations, structural variations, Copy number variations, RNA sequencing, Whole-genome sequencing
Address and Contact Information 1 Department of Nuclear Medicine, Xinhua Hospital Afliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
2 Department of Nuclear Medicine, Shanxi Bethune Hospital (Shanxi Academy of Medical Sciences), Taiyuan 03003, China.
3 Department of Oncology, Xin Hua Hospital Afliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
4 Department of Respiratory Medicine, XinHua Hospital Afliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
5 BioGenius Bioinformatics Institute, Shanghai 200050, People’s Republic of China.
6 Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
7 Medical Center on Aging of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
* Corresponding author: michael@biogenius.cn; liangsheng364214@163.com; lei.huang@alumni.dkfz.de; huiwangshanghai@163.com
Linlin Zhang, Miaomiao Xu, and Wanchun Zhang contributed equally to this work.
Read full article at BMC

No.02  DOI: 10.1186/s11658-022-00413-w Volume 28 (2023) - 28:02
Title Robo4 INHIBITS GAMMA RADIATION-INDUCED PERMEABILITY OF A MURINE MICROVASCULAR ENDOTHELIAL CELL BY REGULATING THE JUNCTIONS
Authors Seyram Yao Adzraku1,2,3,4†, Guozhang Wang1,2,3,4†, Can Cao1,2,3,4†, Yurong Bao1,2,3, Yizhou Wang1,2,3, Alhaji Osman Smith1,2,3, Yuwei Du1,2,3, Haiyang Wang1,2,3, Yue Li1,2,3, Kailin Xu1,2,3, Jianlin Qiao1,2,3*, Wen Ju1,2,3,4* and Lingyu Zeng1,2,3*
Abstract Background: Hematopoietic stem cell transplantation involves irradiation preconditioning which causes bone marrow endothelial cell dysfunction. While much emphasis is on the reconstitution of hematopoietic stem cells in the bone marrow microenvironment, endothelial cell preservation is indispensable to overcome the preconditioning damages. This study aims to ascertain the role of Roundabout 4 (Robo4) in regulating irradiation-induced damage to the endothelium.
Methods: Microvascular endothelial cells were treated with γ-radiation to establish an endothelial cell injury model. Robo4 expression in the endothelial cells was manipulated employing lentiviral-mediated RNAi and gene overexpression technology before irradiation treatment. The permeability of endothelial cells was measured using qPCR, immunocytochemistry, and immunoblotting to analyze the efect on the expression and distribution of junctional molecules, adherens junctions, tight junctions, and gap junctions. Using Transwell endothelial monolayer staining, FITC-Dextran permeability, and gap junction-mediated intercellular communication (GJIC) assays, we determined the changes in endothelial functions after Robo4 gene manipulation and irradiation. Moreover, we measured the proportion of CD31 expression in endothelial cells by fow cytometry. We analyzed variations between two or multiple groups using Student’s t-tests and ANOVA.
Results: Ionizing radiation upregulates Robo4 expression but disrupts endothelial junctional molecules. Robo4 deletion causes further degradation of endothelial junctions hence increasing the permeability of the endothelial cell monolayer. Robo4 knockdown in microvascular endothelial cells increases the degradation and delocalization of ZO-1, PECAM-1, occludin, and claudin-5 molecules after irradiation. Conversely, connexin 43 expression increases after silencing Robo4 in endothelial cells to induce permeability but are readily destroyed when exposed to 10 Gy of gamma radiation. Also, Robo4 knockdown enhances Y731-VE-cadherin phosphorylation leading to the depletion and destabilization of VE-cadherin at the endothelial junctions following irradiation. However, Robo4 overexpression mitigates irradiation-induced degradation of tight junctional proteins and stabilizes claudin-5 and ZO-1 distribution. Finally, the enhanced expression of Robo4 ameliorates the irradiation-induced depletion of VE-cadherin and connexin 43, improves the integrity of microvascular endothelial cell junctions, and decreases permeability.
Conclusion: This study reveals that Robo4 maintains microvascular integrity after radiation preconditioning treatment by regulating endothelial permeability and protecting endothelial functions. Our results also provided a potential mechanism to repair the bone marrow vascular niche after irradiation by modulating Robo4 expression.
Keywords Robo4, Microvascular endothelial cells, Endothelial junctions, Irradiation, Permeability
Address and Contact Information 1 Blood Diseases Institute, Xuzhou Medical University, Xuzhou 221002, China
2 Key Laboratory of Bone Marrow Stem Cell, Xuzhou 221002, Jiangsu, China
3 Department of Hematology, The Afliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
4 Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou 221002, China
*Corresponding author: jlqiao85@126.com; juwen1987-10@163.com; zengly2000@163.com
Seyram Yao Adzraku, Guozhang Wang and Can Cao have contributed equally to this work
Read full article at BMC

No. 03 DOI: 10.1186/s11658-023-00419-y Volume 28 (2023) - 28:03
Title CORRECTION: TMEM43 PROMOTES PANCREATIC CANCER PROGRESSION BY STABILIZING PRPF3 AND REGULATING RAP2B/ERK AXIS
Authors Junqiang Li1†, Yang Song, Chao Zhang1†, Ronglin Wang1, Lei Hua1, Yongdong Guo, Dongxue Gan1, Liaoliao Zhu1, Shanshan Li1, Peixiang Ma1, Cheng Yang1, Hong Li1, Jing Yang1, Jingjie Shi1, Xiaonan Liu2* and Haichuan Su1*
Abstract Correction: Cellular & Molecular Biology Letters (2022) 27:24
https://doi.org/10.1186/s11658-022-00321-z.
Following publication of the original article [1], the authors identifed an error in Fig. 3O. Te invasion image in the shTMEM43+RAP2B group of Fig. 3O was inadvertently placed by mistake. We have double checked the original data and found that the inadvertent errors occurred during picture compilation, and this correction does not change the scientifc conclusions of the article. Te incorrect and the correct figure is given in PDF below.
The online version of the original article can be found at https://doi.org/10.1186/s11658-022-00321-z.
Address and Contact Information 1 Department of Oncology, Tangdu Hospital, Air Force Medical University, Xi’an 710038, Shaanxi, China
2 Ambulatory Surgery Center, Xijing Hospital, Air Force Medical University, Xi’an 710032, Shaanxi, China
*Corresponding author: 15353589999@163.com; suhc@fmmu.edu.cn
Junqiang Li, Yang Song and Chao Zhang contributed equally
Read full article at BMC

No. 04 DOI: 10.1186/s11658-023-00416-1 Volume 28 (2023) - 28:04
Title FORMYL PEPTIDE RECEPTOR 2 ACTIVATION BY MITOCHONDRIAL FORMYL PEPTIDES STIMULATES THE NEUTROPHIL PROINFAMMATORY RESPONSE VIA THE ERK PATHWAY AND EXACERBATES ISCHEMIA–REPERFUSION INJURY
Authors Yirui Cao1,2†, Juntao Chen1,2†, Feng Liu4†, Guisheng Qi1, Yufeng Zhao1,2, Shihao Xu1,2, Jiyan Wang1,2, Tongyu Zhu1,2*†, Yi Zhang3*† and Yichen Jia1*†
Abstract Background: Ischemia–reperfusion injury (IRI) is an inevitable process in renal transplantation that signifcantly increases the risk of delayed graft function, acute rejection, and even graft loss. Formyl peptide receptor 2 (FPR2) is an important receptor in multiple septic and aseptic injuries, but its functions in kidney IRI are still unclear. This study was designed to reveal the pathological role of FPR2 in kidney IRI and its functional mechanisms.
Methods: To explore the mechanism of FPR2 in kidney IRI, the model rats were sacrifced after IRI surgery. Immunofuorescence, enzyme-linked immunosorbent assays, and western blotting were used to detect diferences in the expression of FPR2 and its ligands between the IRI and control groups. WRW4 (WRWWWW-NH2), a specifc antagonist of FPR2, was administered to kidney IRI rats. Kidney function and pathological damage were detected to assess kidney injury and recovery. Flow cytometry was used to quantitatively compare neutrophil infltration among the experimental groups. Mitochondrial formyl peptides (mtFPs) were synthesized and administered to primary rat neutrophils together with the specifc FPR family antagonist WRW4 to verify our hypothesis in vitro. Western blotting and cell function assays were used to examine the functions and signaling pathways that FPR2 mediates in neutrophils.
Results: FPR2 was activated mainly by mtFPs during the acute phase of IRI, mediating neutrophil migration and reactive oxygen species production in the rat kidney through the ERK1/2 pathway. FPR2 blockade in the early phase protected rat kidneys from IRI.
Conclusions: mtFPs activated FPR2 during the acute phase of IRI and mediated rat kidney injury by activating the migration and reactive oxygen species generation of neutrophils through the ERK1/2 pathway.
Keywords Formyl peptide receptor 2, Neutrophil, Kidney IRI, Migration, Mitochondrial-derived formyl peptides
Address and Contact Information 1 Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
2 Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
3 Zhongshan Hospital Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
4 Department of Integrative Medicine, Huashan Hospital Fudan University, Shanghai, People’s Republic of China
* Correspondence: zs_tyzhu@163.com; yzhang_med@fudan.edu.cn; jia.yichen@zs-hospital.sh.cn
Yirui Cao, Juntao Chen, and Feng Liu contributed equally to this work and share first authorship
Tongyu Zhu, Yi Zhang, and Yichen Jia share last authorship
Read full article at BMC

No. 05 DOI: 10.1186/s11658-023-00417-0 Volume 28 (2023) - 28:05
Title PHOTOBIOMODULATION PROMOTES SPINAL CORD INJURY REPAIR BY INHIBITING MACROPHAGE POLARIZATION THROUGH lncRNA TUG1-miR-1192/TLR3 AXIS
Authors Cheng Ju, Yangguang Ma, Xiaoshuang Zuo, Xuankang Wang, Zhiwen Song, Zhihao Zhang, Zhijie Zhu, Xin Li, Zhuowen Liang, Tan Ding, Xueyu Hu* and Zhe Wang*
Abstract Background: Secondary spinal cord injury (SCI) often causes the aggravation of infammatory reaction and nerve injury, which afects the recovery of motor function. Bone-marrow-derived macrophages (BMDMs) were recruited to the injured area after SCI, and the M1 polarization is the key process for inducing infammatory response and neuronal apoptosis. We previously showed that photobiomodulation (PBM) can inhibit the polarization of M1 phenotype of BMDMs and reduce infammation, but the underlying mechanisms are unclear. The purpose of this study is to explore the potential target and mechanism of PBM in treating SCI.
Methods: Transcriptome sequencing and bioinformatics analysis showed that long noncoding RNA taurine upregulated gene 1 (lncRNA TUG1) was a potential target of PBM. The expression and specifc mechanism of lncRNA TUG1 were detected by qPCR, immunofuorescence, fow cytometry, western blotting, fuorescence in situ hybridization, and luciferase assay. The Basso mouse scale (BMS) and gait analysis were used to evaluate the recovery of motor function in mice.
Results: Results showed that lncRNA TUG1 may be a potential target of PBM, regulating the polarization of BMDMs, infammatory response, and the axial growth of DRG. Mechanistically, TUG1 competed with TLR3 for binding to miR-1192 and attenuated the inhibitory efect of miR-1192 on TLR3. This efect protected TLR3 from degradation, enabling the high expression of TLR3, which promoted the activation of downstream NF-κB signal and the release of infammatory cytokines. In vivo, PBM treatment could reduce the expression of TUG1, TLR3, and infammatory cytokines and promoted nerve survival and motor function recovery in SCI mice.
Conclusions: Our study clarifed that the lncRNA TUG1/miR-1192/TLR3 axis is an important pathway for PBM to inhibit M1 macrophage polarization and infammation, which provides theoretical support for its clinical application in patients with SCI.
Keywords Spinal cord injury, Bone-marrow-derived macrophages, Photobiomodulation, Transcriptome sequencing, Infammation, Long noncoding RNA TUG1
Address and Contact Information Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Changle West Road No. 127, Xi’an 710032, Shaanxi, China
*Corresponding author: huxueyu@fmmu.edu.cn; wangzhe@fmmu.edu.cn
Cheng Ju, Yangguang Ma, and Xiaoshuang Zuo have contributed equally to this work and share first authorship
Read full article at BMC

No. 06 DOI: 10.1186/s11658-023-00418-z Volume 28 (2023) - 28:06
Title TRIM8: A DOUBLE-EDGED SWORD IN GLIOBLASTOMA WITH THE POWER TO HEAL OR HURT
Authors Hamed Hosseinalizadeh1, Omid Mohamadzadeh2, Mohammad Saeed Kahrizi3, Zahra Razaghi Bahabadi4,5, Daniel J. Klionsky6 and Hamed Mirzei7*
Abstract Glioblastoma multiforme (GBM) is an aggressive primary brain tumor and one of the most lethal central nervous system tumors in adults. Despite signifcant breakthroughs in standard treatment, only about 5% of patients survive 5 years or longer. Therefore, much efort has been put into the search for identifying new glioma-associated genes. Tripartite motif-containing (TRIM) family proteins are essential regulators of carcino-genesis. TRIM8, a member of the TRIM superfamily, is abnormally expressed in high-grade gliomas and is associated with poor clinical prognosis in patients with glioma. Recent research has shown that TRIM8 is a molecule of duality (MoD) that can function as both an oncogene and a tumor suppressor gene, making it a “double-edged sword” in glioblastoma development. This characteristic is due to its role in selectively regulating three major cellular signaling pathways: the TP53/p53-mediated tumor suppression pathway, NFKB/NF-κB, and the JAK-STAT pathway essential for stem cell property support in glioma stem cells. In this review, TRIM8 is analyzed in detail in the context of GBM and its involvement in essential signaling and stem cell-related pathways. We also discuss the basic biological activities of TRIM8 in macroautophagy/autophagy, regulation of bipolar spindle formation and chromosomal stability, and regulation of chemoresistance, and as a trigger of infammation.
Keywords Autophagy, Glioblastoma, JAK-STAT, NF-κB, p53, Stem-cell, TRIM8
Address and Contact Information 1 Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
2 Department of Neurosurgery, Tehran University of Medical Science, Tehran, Iran
3 Department of Surgery, Alborz University of Medical Sciences, Karaj, Alborz, Iran
4 School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
5 Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
6 Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
7 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
*Corresponding author: mirzaei-h@kaums.ac.ir; h.mirzaei2002@gmail.com
Read full article at BMC

No. 07 DOI: 10.1186/s11658-023-00420-5 Volume 28 (2023) - 28:07
Title LRP6/FLAMENTOUS-ACTIN SIGNALING FACILITATES OSTEOGENIC COMMITMENT IN MECHANICALLY INDUCED PERIODONTAL LIGAMENT STEM CELLS
Authors Jixiao Wang, Huiqi Yang, Xiaobei Ma, Jiani Liu, Lan Li, Lei Chen* and Fulan Wei*
Abstract Background: Mechanotransduction mechanisms whereby periodontal ligament stem cells (PDLSCs) translate mechanical stress into biochemical signals and thereby trigger osteogenic programs necessary for alveolar bone remodeling are being deciphered. Low-density lipoprotein receptor-related protein 6 (LRP6), a Wnt transmembrane receptor, has been qualifed as a key monitor for mechanical cues. However, the role of LRP6 in the mechanotransduction of mechanically induced PDLSCs remains obscure.
Methods: The Tension System and tooth movement model were established to determine the expression profle of LRP6. The loss-of-function assay was used to investigate the role of LRP6 on force-regulated osteogenic commitment in PDLSCs. The ability of osteogenic diferentiation and proliferation was estimated by alkaline phosphatase (ALP) staining, ALP activity assay, western blotting, quantitative real-time PCR (qRT-PCR), and immunofuorescence. Crystalline violet staining was used to visualize cell morphological change. Western blotting, qRT-PCR, and phalloidin staining were adopted to afrm flamentous actin (F-actin) alteration. YAP nucleoplasmic localization was assessed by immunofuorescence and western blotting. YAP transcriptional response was evaluated by qRT-PCR. Cytochalasin D was used to determine the efects of F-actin on osteogenic commitment and YAP switch behavior in mechanically induced PDLSCs.
Results: LRP6 was robustly activated in mechanically induced PDLSCs and PDL tissues. LRP6 defciency impeded force-dependent osteogenic diferentiation and proliferation in PDLSCs. Intriguingly, LRP6 loss caused cell morphological aberration, F-actin dynamics disruption, YAP nucleoplasmic relocation, and subsequent YAP inactivation. Moreover, disrupted F-actin dynamics inhibited osteogenic diferentiation, proliferation, YAP nuclear translocation, and YAP activation in mechanically induced PDLSCs.
Conclusions: We identifed that LRP6 in PDLSCs acted as the mechanosensor regulating mechanical stress-inducible osteogenic commitment via the F-actin/YAP cascade. Targeting LRP6 for controlling alveolar bone remodeling may be a prospective therapy to attenuate relapse of orthodontic treatment.
Keywords Mechanical stress, PDLSCs, Mechanotransduction, LRP6, Filamentous actin, Osteogenic commitment
Address and Contact Information Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, No. 44-1 Wenhua Road West, Jinan 250012, Shandong, China
*Corresponding author: sdcl8006@163.com; weif@email.sdu.edu.cn
Read full article at BMC

No. 08 DOI: 10.1186/s11658-023-00421-4 Volume 28 (2023) - 28:08
Title OUTSIDE THE LIMIT: QUESTIONING THE DISTANCE RESTRICTIONS FOR COOPERATIVE miRNA BINDING SITES
Authors Caroline Diener1*, Martin Hart1, Claudia Fecher‐Trost2, Jessica Knittel1, Stefanie Rheinheimer1, Markus R. Meyer3, Jens Mayer1, Veit Flockerzi2, Andreas Keller4,5 and Eckart Meese1
Abstract Among the concepts in biology that are widely taken granted is a potentiated cooperative efect of multiple miRNAs on the same target. This strong hypothesis contrasts insufcient experimental evidence. The quantity as well as the quality of required side constraints of cooperative binding remain largely hidden. For miR-21-5p and miR-155-5p, two commonly investigated regulators across diseases, we selected 15 joint target genes. These were chosen to represent various neighboring 3′UTR binding site constellations, partially exceeding the distance rules that have been established for over a decade. We identifed diferent cooperative scenarios with the binding of one miRNA enhancing the binding efects of the other miRNA and vice versa. Using both, reporter assays and whole proteome analyses, we observed these cooperative miRNA efects for genes that bear 3′UTR binding sites at distances greater than the previously defned limits. Astonishingly, the experiments provide even stronger evidence for cooperative miRNA efects than originally postulated. In the light of these fndings the defnition of targetomes specifed for single miRNAs need to be refned by a concept that acknowledges the cooperative efects of miRNAs.
Keywords microRNA, Cooperativity, Binding sites, Distances, In silico prediction, RISC, Functional interactions, Networks, Proteomics, Target identifcation
Address and Contact Information 1 Institute of Human Genetics, Saarland University, 66421 Homburg, Germany
2 Department of Experimental and Clinical Pharmacology & Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany
3 Department of Experimental and Clinical Toxicology & Pharmacology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany
4 Chair for Clinical Bioinformatics, Saarland Informatics Campus, Saarland University, 66123 Saarbrücken, Germany
5 Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), 66123 Saarbrücken, Germany
*Corresponding author: caroline.diener@uni-saarland.de
Read full article at BMC

No. 09 DOI:10.1186/s11658-023-00422-3 Volume 28 (2023) - 28:09
Title LOW‐INTENSITY PULSED ULTRASOUND (LIPUS) ENHANCES THE ANTI‐INFAMMATORY EFECTS OF BONE MARROW MESENCHYMAL STEM CELLS (BMSCs)‐DERIVED EXTRACELLULAR VESICLEs
Authors Xueke Li1,2,3†, Yi Zhong1,2,3†, Wuqi Zhou1,2,3, Yishu Song1,2,3, Wenqu Li1,2,3, Qiaofeng Jin1,2,3, Tang Gao1,2,3, Li Zhang1,2,3* and Mingxing Xie1,2,3*
Abstract Background: Bone marrow-derived mesenchymal stem cells (BMSCs)-derived extracellular vesicles (EVs) have shown potent anti-infammatory function in various pathological conditions, such as osteoarthritis and neurodegenerative diseases. Since the number of EVs naturally secreted by cells is fnite and they usually bear specifc repertoires of bioactive molecules to perform manifold cell–cell communication, but not one particular therapeutic function as expected, their practical application is still limited. Strategies are needed to increase the production of EVs and enhance their therapeutic function. Recent studies have suggested that low-intensity pulsed ultrasound (LIPUS) is a promising non-invasive method to increase the secretion of EVs and promote their anti-infammatory efects. However, the efect of LIPUS stimulation of BMSCs on EVs derived from the cells remains unclear. The objective of this study was to investigate whether LIPUS stimulation on BMSCs could increase the secretion of EVs and enhance their anti-infammatory efects.
Methods: BMSCs were exposed to LIPUS (300 mW/cm2) for 15 min and EVs were isolated by ultracentrifugation. Anti-infammatory efects of EVs were investigated on RAW264.7 cells in vitro and in the allogeneic skin transplantation model. Small RNA-seq was utilized to identify components diference in EVs with/without LIPUS irradiation.
Results: In this study, we found that LIPUS stimulation could lead to a 3.66-fold increase in the EVs release from BMSCs. Moreover, both in vitro and in vivo experimental results suggested that EVs secreted from LIPUS-treated BMSCs (LIPUS-EVs) possessed stronger anti-infammatory function than EVs secreted from BMSCs without LIPUS stimulation (C-EVs). RNA-seq analysis revealed that miR-328-5p and miR-487b-3p were signifcantly up-regulated in LIPUS-EVs compare with C-EVs. The suppression of MAPK signaling pathway by these two up-regulated miRNAs could be the potential mechanism of strengthened anti-infammatory efects of LIPUS-EVs.
Conclusion: LIPUS stimulation on BMSCs could signifcantly increase the secretion of EVs. Moreover, EVs generated from LIPUS-treated BMSCs possessed much stronger antiinfammatory function than C-EVs. Therefore, LIPUS could be a promising non-invasive strategy to promote the production of EVs from BMSCs and augment their anti-infammatory efects.
Keywords Low-intensity pulsed ultrasound, Extracellular vesicles, Infammation, Bone marrow mesenchymal stem cells, microRNA
Address and Contact Information 1 Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
2 Clinical Research Center for Medical Imaging in Hubei Province, Wuhan 430022, China
3 Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
*Correspondence: zli429@hust.edu.cn; xiemx@hust.edu.cn
Xueke Li and Yi Zhong have equally contributed to this work
Read full article at BMC

No. 10 DOI: 10.1186/s11658-023-00424-1 Volume 28 (2023) - 28:10
Title A THERAPEUTIC TARGET FOR CKD: ACTIVIN A FACILITATES TGFΒ1 PROFBROTIC SIGNALING
Authors Asfa Soomro1, Mohammad Khajehei1, Renzhong Li1, Kian O’Neil1, Dan Zhang1, Bo Gao1, Melissa MacDonald1, Masao Kakoki2 and Joan C. Krepinsky1,3*
Abstract Background: TGFβ1 is a major profbrotic mediator in chronic kidney disease (CKD). Its direct inhibition, however, is limited by adverse efects. Inhibition of activins, also members of the TGFβ superfamily, blocks TGFβ1 profbrotic efects, but the mechanism underlying this and the specifc activin(s) involved are unknown.
Methods: Cells were treated with TGFβ1 or activins A/B. Activins were inhibited generally with follistatin, or specifcally with neutralizing antibodies or type I receptor downregulation. Cytokine levels, signaling and profbrotic responses were assessed with ELISA, immunofuorescence, immunoblotting and promoter luciferase reporters. Wild-type or TGFβ1-overexpressing mice with unilateral ureteral obstruction (UUO) were treated with an activin A neutralizing antibody.
Results: In primary mesangial cells, TGFβ1 induces secretion primarily of activin A, which enables longer-term profbrotic efects by enhancing Smad3 phosphorylation and transcriptional activity. This results from lack of cell refractoriness to activin A, unlike that for TGFβ1, and promotion of TGFβ type II receptor expression. Activin A also supports transcription through regulating non-canonical MRTF-A activation. TGFβ1 additionally induces secretion of activin A, but not B, from tubular cells, and activin A neutralization prevents the TGFβ1 profbrotic response in renal fbroblasts. Fibrosis induced by UUO is inhibited by activin A neutralization in wild-type mice. Worsened fbrosis in TGFβ1-overexpressing mice is associated with increased renal activin A expression and is inhibited to wild-type levels with activin A neutralization.
Conclusions: Activin A facilitates TGFβ1 profbrotic efects through regulation of both canonical (Smad3) and non-canonical (MRTF-A) signaling, suggesting it may be a novel therapeutic target for preventing fbrosis in CKD.
Keywords Activin A, TGFβ1, Kidney fbrosis, Extracellular matrix
Address and Contact Information 1 Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
2 Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
3 St. Joseph’s Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON L8N 4A6, Canada
*Corresponding author: krepinj@mcmaster.ca
Read full article at BMC

No. 11 DOI: 10.1186/s11658-023-00426-z Volume 28 (2023) - 28:11
Title SINGLE-CELL TRANSCRIPTOME ANALYSIS REVEALS LIVER INJURY INDUCED BY GLYPHOSATE IN MICE
Authors Jiangpeng Wu1†, Xiuping Sun1†, Chunyi Wu1†, Xiaoping Hong2†, Lulin Xie1, Zixu Shiv1, Liang Zhao6,7, Qingfeng Du4*, Wei Xiao3,5*, Jichao Sun1* and Jigang Wang1,4,5,8*
Abstract Background: Glyphosate (GLY), as the active ingredient of the most widely used herbicide worldwide, is commonly detected in the environment and living organisms, including humans. Its toxicity and carcinogenicity in mammals remain controversial. Several studies have demonstrated the hepatotoxicity of GLY; however, the underlying cellular and molecular mechanisms are still largely unknown.
Methods: Using single-cell RNA sequencing (scRNA-seq), immunofuorescent staining, and in vivo animal studies, we analyzed the liver tissues from untreated and GLY-treated mice.
Results: We generated the frst scRNA-seq atlas of GLY-exposed mouse liver. GLY induced varied cell composition, shared or cell-type-specifc transcriptional alterations, and dysregulated cell–cell communication and thus exerted hepatotoxicity efects. The oxidative stress and infammatory response were commonly upregulated in several cell types. We also observed activation and upregulated phagocytosis in macrophages, as well as proliferation and extracellular matrix overproduction in hepatic stellate cells.
Conclusions: Our study provides a comprehensive single-cell transcriptional picture of the toxic efect of GLY in the liver, which ofers novel insights into the molecular mechanisms of the GLY-associated hepatotoxicity.
Keywords Glyphosate, Hepatotoxicity, scRNA-seq
Address and Contact Information 1 Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Afliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
2 Department of Rheumatology and Immunology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Afliated Hospital, Southern University of Science and Technology), Shenzhen 518020, China.
3 Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, China.
4 School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
5 Dongguan Maternal and Child Health Care Hospital, Postdoctoral Innovation Practice Base of Southern Medical University, Dongguan 523125, Guangdong, China.
6 Department of Pathology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde), Foshan 528300, China.
7 Department of Pathology and Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
8 Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
*Corresponding author: dqfsmu7@126.com; xw7688@smu.edu.cn; sunjichao@mail.sustech.edu.cn; jgwang@icmm.ac.cn
Jiangpeng Wu, Xiuping Sun, Chunyi Wu, and Xiaoping Hong contributed equally to this work.
Read full article at BMC

No.  12DOI: 10.1186/s11658-023-00425-0 Volume 28 (2023) - 28:12
Title HUMAN UMBILICAL CORD MESENCHYMAL STEM CELL EXOSOME-DERIVED miR-874-3p TARGETING RIPK1/PGAM5 ATTENUATES KIDNEY TUBULAR EPITHELIAL CELL DAMAGE
Authors Yihang Yu1,2,3, Meiling Chen1,2,3, Qitong Guo1,2,3, Lianju Shen1,2,3, Xing Liu1,2,3, Jianbo Pan4, Yuanyuan Zhang5, Tao Xu6, Deying Zhang1,2,3* and Guanghui Wei1,2,3
Abstract Background: Kidney insults due to various pathogenic factors, such as trauma, infection, and infammation, can cause tubular epithelial cell injury and death, leading to acute kidney injury and the transformation of acute kidney injury to chronic kidney disease. There is no defnitive treatment available. In previous studies, human umbilical cord mesenchymal stem cells have been shown to promote kidney injury. In this preclinical study, we investigate the role and mechanism of human umbilical cord mesenchymal stem cell exosomes (HucMSC-Exos) on the repair of renal tubular epithelial cells after injury.
Methods: C57BL/6 mice underwent unilateral ureteral obstruction, and epithelial cell injury was induced in HK-2 cells by cisplatin. HucMSC-Exos were assessed in vivo and in vitro. The extent of renal cell injury, activation of necroptosis pathway, and mitochondrial quality-control-related factors were determined in diferent groups. We also analyzed the possible regulatory efector molecules in HucMSC-Exos by transcriptomics.
Results: HucMSC-Exo inhibited necroptosis after renal tubular epithelial cell injury and promoted the dephosphorylation of the S637 site of the Drp1 gene by reducing the expression of PGAM5. This subsequently inhibited mitochondrial fission and maintained mitochondrial functional homeostasis, mitigating renal injury and promoting repair. In addition, HucMSC-Exo displayed a regulatory role by targeting RIPK1 through miR-874-3p.
Conclusion: The collective findings of the present study demonstrate that HucMSC-Exos can regulate necroptosis through miR-874-3p to attenuate renal tubular epithelial cell injury and enhance repair, providing new therapeutic modalities and ideas for the treatment of AKI and the process of AKI to CKD transformation to mitigate renal damage.
Keywords Human umbilical cord mesenchymal stem cells, Exosome, Necroptosis, Mitochondrial fusion, miR-874-3p, Kidney tubular epithelial cell damage
Address and Contact Information 1 Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
2 Chongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing 400014, China
3 National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
4 Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
5 Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
6 Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
*Corresponding author: zdy@hospital.cqmu.edu.cn
Read full article at BMC

No.  13DOI: 10.1186/s11658-022-00410-z Volume 28 (2023) - 28:13
Title EXOSOMAL circ_0026611 CONTRIBUTES TO LYMPHANGIOGENESIS BY REDUCING PROX1 ACETYLATION AND UBIQUITINATION IN HUMAN LYMPHATIC ENDOTHELIAL CELLS (HLECs)
Authors Wenjian Yao1†, Xiangbo Jia1†, Li Zhu2, Lei Xu1, Quan Zhang1, Tian Xia1 and Li Wei1*
Abstract Background: Esophageal squamous carcinoma (ESCC) is a common malignancy that originates in the digestive tract. Lymph node metastasis (LNM) is a complicated process, and tumor lymphangiogenesis has been reported to be associated with the spread of tumor cells to lymph nodes (LNs), including in ESCC. However, little is currently known about the mechanisms involved in lymphangiogenesis in ESCC tumors. According to previous literature, we know that hsa_circ_0026611 expresses at a high level in serum exosomes of patients with ESCC and shows a close association with LNM and poor prognosis. However, details on the functions of circ_0026611 in ESCC remain unclear. We aim to explore the efects of circ_0026611 in ESCC cell-derived exosomes on lymphangiogenesis and its potential molecular mechanism.
Methods: We firstly examined how circ_0026611 may express in ESCC cells and exosomes by quantitative reverse transcription real-time polymerase chain reaction (RT-qPCR). The potential efects circ_0026611 may exert on lymphangiogenesis in ESCC cell-derived exosomes were assessed afterward via mechanism experiments.
Results: circ_0026611 high expression pattern was confrmed in ESCC cells and exosomes. ESCC cell-derived exosomes promoted lymphangiogenesis by transferring circ_0026611. Besides, circ_0026611 interacted with N-α-acetyltransferase 10 (NAA10) to inhibit NAA10-mediated prospero homeobox 1 (PROX1) acetylation with subsequent ubiquitination and degradation. Furthermore, circ_0026611 was verifed to promote lymphangiogenesis in a PROX1-mediated manner.
Conclusions: Exosomal circ_0026611 inhibited PROX1 acetylation and ubiquitination to promote lymphangiogenesis in ESCC.
Keywords Esophageal squamous cell carcinoma, Exosomes, circ_0026611, PROX1, Lymphangiogenesis
Address and Contact Information 1 Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, No.7, Weiwu Road, Jinshui District, Zhengzhou 450003, Henan, China
2 Department of Thoracic Surgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou 450003, Henan, China
*Corresponding author: zhuwei627328429@163.com
Wenjian Yao and Xiangbo Jia co-first authors.
Read full article at BMC

No.  14DOI: 10.1186/s11658-023-00427-y Volume 28 (2023) - 28:14
Title PATTERNS OF HUMAN AND PORCINE GAMMAHERPESVIRUS-ENCODED BILF1 RECEPTOR ENDOCYTOSIS
Authors Maša Mavri1, Sanja Glišić2, Milan Senćanski2, Milka Vrecl1, Mette M. Rosenkilde3, Katja Spiess3,4 and Valentina Kubale1*
Abstract Background: The viral G-protein-coupled receptor (vGPCR) BILF1 encoded by the Epstein–Barr virus (EBV) is an oncogene and immunoevasin and can downregulate MHC-I molecules at the surface of infected cells. MHC-I downregulation, which presumably occurs through co-internalization with EBV-BILF1, is preserved among BILF1 receptors, including the three BILF1 orthologs encoded by porcine lymphotropic herpesviruses (PLHV BILFs). This study aimed to understand the detailed mechanisms of BILF1 receptor constitutive internalization, to explore the translational potential of PLHV BILFs compared with EBV-BILF1.
Methods: A novel real-time fluorescence resonance energy transfer (FRET)-based internalization assay combined with dominant-negative variants of dynamin-1 (Dyn K44A) and the chemical clathrin inhibitor Pitstop2 in HEK-293A cells was used to study the effect of specific endocytic proteins on BILF1 internalization. Bioluminescence resonance energy transfer (BRET)-saturation analysis was used to study BILF1 receptor interaction with β-arrestin2 and Rab7. In addition, a bioinformatics approach informational spectrum method (ISM) was used to investigate the interaction affinity of BILF1 receptors with β-arrestin2, AP-2, and caveolin-1.
Results: We identified dynamin-dependent, clathrin-mediated constitutive endocytosis for all BILF1 receptors. The observed interaction affinity between BILF1 receptors and caveolin-1 and the decreased internalization in the presence of a dominant-negative variant of caveolin-1 (Cav S80E) indicated the involvement of caveolin-1 in BILF1 trafficking. Furthermore, after BILF1 internalization from the plasma membrane, both the recycling and degradation pathways are proposed for BILF1 receptors.
Conclusions: The similarity in the internalization mechanisms observed for EBV-BILF1 and PLHV1-2 BILF1 provide a foundation for further studies exploring a possible translational potential for PLHVs, as proposed previously, and provides new information about receptor trafficking.
Keywords vGPCR, BILF1, Endocytosis, Internalization, Dynamin, Caveolin, β-Arrestin, EBV, PLHV1-2
Address and Contact Information 1 Institute for preclinical sciences, Veterinary Faculty, Ljubljana, Slovenia
2 Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
3 Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
4 Present Address: Department of Virus and Microbiological Special Diagnostics, Statens Serum Institute, Copenhagen, Denmark
*Corresponding author: valentina.kubale@vf.uni-lj.si
Read full article at BMC

No.  15DOI: 10.1186/s11658-023-00429-w Volume 28 (2023) - 28:15
Title IRE1α ARM OF UNFOLDED PROTEIN RESPONSE IN MUSCLE-SPECIFC TGF-β SIGNALING-MEDIATED REGULATION OF MUSCLE CELL IMMUNOLOGICAL PROPERTIES
Authors Jiangwei Xiao1†, Jingwen Huang1†, Xiaoting Jian1, Han Wang1, Haiqiang Lan1, Zhaohong Liao1, Ruicai Gu2, Jijie Hu3* and Hua Liao1*
Abstract Endoplasmic reticulum stress (ERS) and the unfolded protein response (UPR) are involved in various muscle pathological states. The IRE1α arm of UPR can afect immunological properties of myofber through restraining p38 mitogen-activated protein kinases (MAPK) activation under infammatory milieu. However, the relevant pathway molecules regulating the initiation of the IRE1α arm in myofiber remain unclear. In this work, expression of transforming growth factor-beta (TGF-β) and TGF-β receptor II (TGF-βr2), and UPR pathway activation were examined in cardiotoxin (CTX)-damaged mouse muscle, which revealed the activation of TGF-β signaling and UPR in CTX-damaged muscle and in regenerating myofibers. Using control or transgenic mice with TGF-βr2 deleted in skeletal muscle (SM TGF-βr2−/−) and the derived primary diferentiating myogenic precursor cells (MPCs) treated with/without ERS activator or inhibitor, IRE1α pathway inhibitor, or TGF-β signaling activator, this study further revealed an essential role of intrinsic TGF-β signaling in regulating muscle cell to express infammation-related molecules including H-2Kb, H2-Eα, TLR3, and special myokines. TGF-β signaling prompted UPR IRE1α arm and restrained p38 MAPK activation in myofiber under infammatory milieu. This study uncovers a previously unrecognized function of TGF-β signaling acting as an upstream factor controlling myofiber immune capacities in the infamed state through the UPR–IRE1α–p38 MAPK pathway.
Keywords Myofiber, Infammation, TGF-β, UPR, IRE1α, p38 MAPK
Address and Contact Information 1 Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
2 Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
3 Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
*Corresponding author: hjj0610@163.com; hua-liao@163.com
Jiangwei Xiao and Jingwen Huang contributed equally to this work
Read full article at BMC

No.  16DOI: 10.1186/s11658-023-00433-0 Volume 28 (2023) - 28:16
Title THE RXFP2-PLC/PKC SIGNALING PATHWAY MEDIATES INSL3-INDUCED REGULATION OF THE PROLIFERATION, MIGRATION AND APOPTOSIS OF MOUSE GUBERNACULAR CELLS
Authors Shouxing Duan1,2, Xuewu Jiang3, Jianhong Li4, Maxian Fu1, Zhuo Li1, Yiyi Cheng2, Yangmu Zhuang2, Ming Yang2, Wenfeng Xiao4, Hongyan Ping3, Yao Xie5* , Xiaojun Xie6* and Xuan Zhang3*
Abstract Background: Testicular hypoplasia can affect the sexual and reproductive ability in adulthood, and even increase the risk of cancer. Abnormal development of the gubernaculum is one of the important factors of testicular hypoplasia. Therefore, a study of the structure and function of the gubernaculum is an important but neglected new breakthrough point for investigating the normal/abnormal development of the testis. Previous findings showed that Insulin like factor 3 (INSL3) is a key factor regulating the growth of gubernaculum, however, the mechanism by which INSL3 acts on the gubernaculum remains unknown. Therefore, we probed the mechanism associated with INSL3-induced the proliferation, migration, and apoptosis of gubernacular cells in mice.
Methods: A culture cell model of neonatal mice gubernaculum is established by INSL3 intervention. We blocked PLC/PKC signaling pathway with U73122 pretreat to investigate the role of the PLC/PKC signaling pathway. The changes of cell proliferation, migration, and apoptosis were detected by molecular biological methods. In addition, the levels of PCNA and F-action were detected by immunofuorescence and western blotting.
Results: We found that INSL3 can promote the proliferation and migration of gubernacular cells and inhibit their apoptosis, meanwhile, INSL3 signifcantly up-regulated PLC/PKC protein phosphorylation. However, treatment with the PLC/PKC signaling pathway inhibitor U73122 significantly inhibited these efects of INSL3. Besides, we found that INSL3 could up-regulate the protein expression level of PCNA and F-actin, while the PCNA and F-actin expression was signifcantly weakened after U73122 pretreatment.
Conclusions: This research revealed that INSL3 binding to RXFP2 may up-regulate the expression levels of PCNA and F-actin by activating the PLC/PKC signaling pathway to promote the proliferation and migration of gubernacular cells. It suggests that the RXFP2-PLC/PKC axis may serve as a novel molecular mechanism by which INSL3 regulates growth of the gubernaculum.
Keywords INSL3, RXFP2, PLC/PKC, Gubernaculum, Signaling pathway
Address and Contact Information 1 Department of Pediatric Surgery, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), No. 89 Taoyuan Road, Shenzhen 518052, Guangdong, China.
2 Department of Pediatric Surgery, The First Afliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou 515041, Guangdong, China.
3 Department of Pediatric Surgery, Pingshan District Maternal and Child Healthcare Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, No. 6 Longxingnan Road, Shenzhen 518118, Guangdong, China.
4 Department of Pediatric Surgery, The Second Afliated Hospital of Shantou University Medical College, No. 69 Dongxiabei Road, Shantou 515041, Guangdong, China.
5 Department of Radiology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515041, Guangdong, China.
6 Department of General Surgery, The First Afliated Hospital of Shantou University Medical College, No. 57 Changping Road, Shantou 515041, Guangdong, China.
*Corresponding author: stxieyao@163.com; xjxie79@163.com; zhxuan2006@126.com
Read full article at BMC

No.  17DOI: 10.1186/s11658-023-00431-2 Volume 28 (2023) - 28:17
Title SYNAPTIC RETROGRADE REGULATION OF THE PKA-INDUCED SNAP-25 AND Synapsin-1 PHOSPHORYLATION
Authors Aleksandra Polishchuk, Víctor Cilleros‐Mañé, Laia Just‐Borràs, Marta Balanyà‐Segura, Genís Vandellòs Pont, Carolina Silvera Simón, Marta Tomàs, Neus Garcia, Josep Tomàs*† and Maria A. Lanuza*†
Abstract Background: Bidirectional communication between presynaptic and postsynaptic components contribute to the homeostasis of the synapse. In the neuromuscular synapse, the arrival of the nerve impulse at the presynaptic terminal triggers the molecular mechanisms associated with ACh release, which can be retrogradely regulated by the resulting muscle contraction. This retrograde regulation, however, has been poorly studied. At the neuromuscular junction (NMJ), protein kinase A (PKA) enhances neurotransmitter release, and the phosphorylation of the molecules of the release machinery including synaptosomal associated protein of 25 kDa (SNAP-25) and Synapsin-1 could be involved.
Methods: Accordingly, to study the effect of synaptic retrograde regulation of the PKA subunits and its activity, we stimulated the rat phrenic nerve (1 Hz, 30 min) resulting or not in contraction (abolished by μ-conotoxin GIIIB). Changes in protein levels and phosphorylation were detected by western blotting and cytosol/membrane translocation by subcellular fractionation. Synapsin-1 was localized in the levator auris longus (LAL) muscle by immunohistochemistry.
Results: Here we show that synaptic PKA Cβ subunit regulated by RIIβ or RIIα subunits controls activity-dependent phosphorylation of SNAP-25 and Synapsin-1, respectively. Muscle contraction retrogradely downregulates presynaptic activity-induced pSynapsin-1 S9 while that enhances pSNAP-25 T138. Both actions could coordinately contribute to decreasing the neurotransmitter release at the NMJ.
Conclusion: This provides a molecular mechanism of the bidirectional communication between nerve terminals and muscle cells to balance the accurate process of ACh release, which could be important to characterize molecules as a therapy for neuromuscular diseases in which neuromuscular crosstalk is impaired.
Keywords Neuromuscular junction, Synapse, Neurotransmission, ACh release, SNAP-25, Synapsin-1, PKA subunits, Synaptic activity, Electrical stimulation
Address and Contact Information Unitat d’Histologia i Neurobiologia (UHNEUROB), Departament de Ciències Mèdiques Bàsiques, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, c/ Sant Llorenç 21, 43201 Reus, Spain
*Corresponding author: josepmaria.tomas@urv.cat; mariaangel.lanuza@urv.cat
Aleksandra Polishchuk and Víctor Cilleros-Mañé contributed equally to this work
Neus Garcia, Josep Tomàs and Maria A. Lanuza contributed equally to this work
Read full article at BMC

No.  18DOI: 10.1186/s11658-023-00432-1 Volume 28 (2023) - 28:18
Title EXOSOMAL Mir-3613-3p DERIVED FROM OXYGEN–GLUCOSE DEPRIVATION-TREATED BRAIN MICROVASCULAR ENDOTHELIAL CELL PROMOTES MICROGLIAL M1 POLARIZATION
Authors Mengqi Zhang1,2*, Qian Wu3, Mimi Tang4,5, Zhuohui Chen1,2 and Haiyue Wu1,2
Abstract Background: Brain microvascular endothelial cell (BMEC) injury can afect neuronal survival by modulating immune responses through the microenvironment. Exosomes are important vehicles of transport between cells. However, the regulation of the subtypes of microglia by BMECs through the exosome transport of microRNAs (miRNAs) has not been established.
Methods: In this study, exosomes from normal and oxygen–glucose deprivation (OGD)-cultured BMECs were collected, and diferentially expressed miRNAs were analyzed. BMEC proliferation, migration, and tube formation were analyzed using MTS, transwell, and tube formation assays. M1 and M2 microglia and apoptosis were analyzed using flow cytometry. miRNA expression was analyzed using real-time polymerase chain reaction (RT-qPCR), and IL-1β, iNOS, IL-6, IL-10, and RC3H1 protein concentrations were analyzed using western blotting.
Results: We found that miR-3613-3p was enriched in BMEC exosome by miRNA GeneChip assay and RT-qPCR analysis. miR-3613-3p knockdown enhanced cell survival, migration, and angiogenesis in the OGD-treated BMECs. In addition, BMECs secrete miR-3613-3p to transfer into microglia via exosomes, and miR-3613-3p binds to the RC3H1 3′ untranslated region (UTR) to reduce RC3H1 protein levels in microglia. Exosomal miR-3613-3p promotes microglial M1 polarization by inhibiting RC3H1 protein levels. BMEC exosomal miR-3613-3p reduces neuronal survival by regulating microglial M1 polarization.
Conclusions: miR-3613-3p knockdown enhances BMEC functions under OGD conditions. Interfering with miR-3613-3p expression in BMSCs reduced the enrichment of miR-3613-3p in exosomes and enhanced M2 polarization of microglia, which contributed to reduced neuronal apoptosis.
Keywords Exosome, Ischemic stroke, Brain microvascular endothelial cell, Macrophage, Neuron
Address and Contact Information 1 Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, China
2 National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
3 Department of Neurology, First Afliated Hospital, Kunming Medical University, Kunming 650032, China
4 Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
5 Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
*Corresponding author: zhangmengqi8912@163.com
Read full article at BMC

No.  19DOI: 10.1186/s11658-023-00428-x Volume 28 (2023) - 28:19
Title 3D GENOMICS AND ITS APPLICATIONS IN PRECISION MEDICINE
Authors Mengjie Chen1, Xingyu Liu1, Qingyou Liu1,2, Deshun Shi1* and Hui Li1*
Abstract Three-dimensional (3D) genomics is an emerging discipline that studies the three-dimensional structure of chromatin and the three-dimensional and functions of genomes. It mainly studies the three-dimensional conformation and functional regulation of intranuclear genomes, such as DNA replication, DNA recombination, genome folding, gene expression regulation, transcription factor regulation mechanism, and the maintenance of three-dimensional conformation of genomes. Self-chromosomal conformation capture (3C) technology has been developed, and 3D genomics and related fields have developed rapidly. In addition, chromatin interaction analysis techniques developed by 3C technologies, such as paired-end tag sequencing (ChIA-PET) and whole-genome chromosome conformation capture (Hi-C), enable scientists to further study the relationship between chromatin conformation and gene regulation in different species. Thus, the spatial conformation of plant, animal, and microbial genomes, transcriptional regulation mechanisms, interaction patterns of chromosomes, and the formation mechanism of spatiotemporal specificity of genomes are revealed. With the help of new experimental technologies, the identifcation of key genes and signal pathways related to life activities and diseases is sustaining the rapid development of life science, agriculture, and medicine. In this paper, the concept and development of 3D genomics and its application in agricultural science, life science, and medicine are introduced, which provides a theoretical basis for the study of biological life processes.
Keywords 3D genomics, Chromatin conformation, Gene expression regulation, Hi-C, Precision biology, Precision medicine
Address and Contact Information 1 State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi Province, China
2 Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, School of Life Science and Engineering, Foshan University, Foshan 528225, China
*Corresponding author: ardsshi@gxu.edu.cn; huili@gxu.edu.cn
Read full article at BMC

No.  20DOI: 10.1186/s11658-023-00430-3 Volume 28 (2023) - 28:20
Title OVEREXPRESSION OF miR-92a ATTENUATES KIDNEY ISCHEMIA–REPERFUSION INJURY AND IMPROVES KIDNEY PRESERVATION BY INHIBITING MEK4/JNK1-RELATED AUTOPHAGY
Authors Ming Ma1,2, Hui Li1,2, Saifu Yin1,2, Tao Lin1,2* and Turun Song1,2*
Abstract Background: Kidney ischemia–reperfusion injury is inevitable in kidney transplantation, and is essential for primary graft dysfunction and delayed graft function. Our previous study has proved that miR-92a could ameliorate kidney ischemia–reperfusion injury, but the mechanism has not been studied.
Methods: This study conducted further research on the role of miR-92a in kidney ischemia–reperfusion injury and organ preservation. In vivo, mice models of bilateral kidney ischemia (30 min), cold preservation after ischemia (cold preservation time of 6, 12, and 24 h), and ischemia–reperfusion (reperfusion time of 24, 48, and 72 h) were established. Before or after modeling, the model mice were injected with miR-92a-ago-mir through the caudal vein. In vitro, the hypoxia–reoxygenation of HK-2 cells was used to simulate ischemia–reperfusion injury.
Results: Kidney ischemia and ischemia–reperfusion signifcantly damaged kidney function, decreased the expression of miR-92a, and increased apoptosis and autophagy in kidneys. miR-92a agomir tail vein injection signifcantly increased the expression of miR-92a in kidneys, improved kidney function, and alleviated kidney injury, and the intervention before modeling achieved a better efect than after. Moreover, miR-92a agomir signifcantly reduced the apoptosis and autophagy in HK-2 cells induced by hypoxia, hypoxia–reoxygenation, and rapamycin, while miR-92a antagomir had opposite efects. Furthermore, mitogen-activated protein kinase, c-Jun NH (2) terminal kinase, caspase 3, Beclin 1, and microtubule-associated protein 1 light chain 3B were inhibited by overexpression of miR-92a both in vivo and in vitro, which in turn reduced apoptosis and autophagy.
Conclusions: Our results prove that overexpression of miR-92a attenuated kidney ischemia–reperfusion injury and improved kidney preservation, and intervention before ischemia–reperfusion provides better protection than after.
Keywords miR-92a, Kidney transplant, Ischemia–reperfusion injury, Autophagy, Apoptosis
Address and Contact Information 1 Department of Urology, West China Hospital, Sichuan University, 37# Guoxue Alley, Chengdu 610041, Sichuan, China
2 Organ Transplantation Center, West China Hospital, Sichuan University, 37# Guoxue Alley, Chengdu 610041, Sichuan, China
*Correspondence: kidney5@163.com; songturun1986@scu.edu.cn
Read full article at BMC

No.  21DOI: 10.1186/s11658-023-00423-2 Volume 28 (2023) - 28:21
Title ENDOTHELIAL DYSFUNCTION DUE TO eNOS UNCOUPLING: MOLECULAR MECHANISMS AS POTENTIAL THERAPEUTIC TARGETS
Authors Anna Janaszak‐Jasiecka1, Agata Płoska1, Joanna M. Wierońska2, Lawrence W. Dobrucki1,3,4,5 and Leszek Kalinowski1,6*
Abstract Nitric oxide (NO) is one of the most important molecules released by endothelial cells, and its antiatherogenic properties support cardiovascular homeostasis. Diminished NO bioavailability is a common hallmark of endothelial dysfunction underlying the pathogenesis of the cardiovascular disease. Vascular NO is synthesized by endothelial nitric oxide synthase (eNOS) from the substrate L-arginine (L-Arg), with tetrahydrobiopterin (BH4) as an essential cofactor. Cardiovascular risk factors such as diabetes, dyslipidemia, hypertension, aging, or smoking increase vascular oxidative stress that strongly affects eNOS activity and leads to eNOS uncoupling. Uncoupled eNOS produces superoxide anion (O2) instead of NO, thus becoming a source of harmful free radicals exacerbating the oxidative stress further. eNOS uncoupling is thought to be one of the major underlying causes of endothelial dysfunction observed in the pathogenesis of vascular diseases. Here, we discuss the main mechanisms of eNOS uncoupling, including oxidative depletion of the critical eNOS cofactor BH4, defciency of eNOS substrate L-Arg, or accumulation of its analog asymmetrical dimethylarginine (ADMA), and eNOS S-glutathionylation. Moreover, potential therapeutic approaches that prevent eNOS uncoupling by improving cofactor availability, restoration of L-Arg/ADMA ratio, or modulation of eNOS S-glutathionylation are briefy outlined.
Keywords Cardiovascular disease, Endothelial dysfunction, eNOS uncoupling, Oxidative/nitroxidative stress, Peroxynitrite, Nitric oxide, ADMA, Tetrahydrobiopterin, BH4
Address and Contact Information 1 Department of Medical Laboratory Diagnostics ‐ Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80‐211, Gdansk, Poland
2 Department of Neurobiology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, 31‐343 Kraków, Poland
3 Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
4 Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC‐251, Urbana, IL 61801, USA
5 Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, Urbana, IL, USA
6 BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Gabriela Narutowicza Street, 80‐233 Gdansk, Poland
*Corresponding author: leszek.kalinowski@gumed.edu.pl
Read full article at BMC

No.  22DOI: 10.1186/s11658-023-00436-x Volume 28 (2023) - 28:22
Title OSCILLATORY SHEAR STRESS MODULATES NOTCH-MEDIATED ENDOTHELIAL MESENCHYMAL PLASTICITY IN CEREBRAL ARTERIOVENOUS MALFORMATIONS
Authors C. L. Karthika1, Vani Venugopal1, B. J. Sreelakshmi1, S. Krithika1, Jaya Mary Thomas1, Mathew Abraham2, C. C. Kartha3, Arumugam Rajavelu4 and S. Sumi1*
ABSTRACT Background: Cerebral arteriovenous malformations (cAVM) are a signifcant cause of intracranial hemorrhagic stroke and brain damage. The arteriovenous junctions in AVM nidus are known to have hemodynamic disturbances such as altered shear stress, which could lead to endothelial dysfunction. The molecular mechanisms coupling shear stress and endothelial dysfunction in cAVMs are poorly understood. We speculated that disturbed blood fow in artery–vein junctions activates Notch receptors and promotes endothelial mesenchymal plasticity during cAVM formation.
Methods: We investigated the expression profle of endothelial mesenchymal transition (EndMT) and cell adhesion markers, as well as activated Notch receptors, in 18 human cAVM samples and 15 control brain tissues, by quantitative real-time PCR (qRT-PCR) and immunohistochemical evaluation. Employing a combination of a microfuidic system, qRT-PCR, immunofuorescence, as well as invasion and inhibitor assays, the efects of various shear stress conditions on Notch-induced EndMT and invasive potential of human cerebral microvascular endothelial cells (hCMEC/d3) were analyzed.
Results: We found evidence for EndMT and enhanced expression of activated Notch intracellular domain (NICD3 and NICD4) in human AVM nidus samples. The expression of transmembrane adhesion receptor integrin α9/β1 is signifcantly reduced in cAVM nidal vessels. Cell–cell adhesion proteins such as VE-cadherin and N-cadherin were diferentially expressed in AVM nidus compared with control brain tissues. Using well-characterized hCMECs, we show that altered fluid shear stress steers Notch3 nuclear translocation and promotes SNAI1/2 expression and nuclear localization. Oscillatory flow downregulates integrin α9/β1 and VE-cadherin expression, while N-cadherin expression and endothelial cell invasiveness are augmented. Gamma-secretase inhibitor RO4929097, and to a lesser level DAPT, prevent the mesenchymal transition and invasiveness of cerebral microvascular endothelial cells exposed to oscillatory fuid fow.
Conclusions: Our study provides, for the first time, evidence for the role of oscillatory shear stress in mediating the EndMT process and dysregulated expression of cell adhesion molecules, especially multifunctional integrin α9/β1 in human cAVM nidus. Concomitantly, our findings indicate the potential use of small-molecular inhibitors such as RO4929097 in the less-invasive therapeutic management of cAVMs.
Keywords Cerebral arteriovenous malformations, Shear stress, Endothelial cells, EndMT, Notch, Small-molecule inhibitors
Address and Contact Information 1 Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala 695014, India
2 Department of Neurosurgery, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala 695011, India
3 Department of Neurology, Amrita Institute of Medical Sciences, Amrita Vishwa Vidyapeetham, Kochi, Kerala 682041, India
4 Department of Biotechnology, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology, Madras, Chennai, Tamil Nadu 600036, India
* Corresponding author: sumis@rgcb.res.in
C. L. Karthika and Vani Venugopal share first authorship
Read full article at BMC

No.  23DOI: 10.1186/s11658-023-00437-w Volume 28 (2023) - 28:23
Title TMEM30A IS ESSENTIAL FOR HAIR CELL POLARITY MAINTENANCE IN POSTNATAL MOUSE COCHLEA
Authors Yazhi Xing1,2†, Kun Peng3†, Qian Yi3, Dongzhen Yu1,2, Haibo Shi1,2, Guang Yang1,2* and Shankai Yin1,2
Abstract Background: Phosphatidylserine is translocated to the inner leaflet of the phospholipid bilayer membrane by the flippase function of type IV P-tape ATPase (P4-ATPase), which is critical to maintain cellular stability and homeostasis. Transmembrane protein 30A (TMEM30A) is the β-subunit of P4-ATPase. Loss of P4-ATPase function causes sensorineural hearing loss and visual dysfunction in human. However, the function of TMEM30A in the auditory system is unclear.
Methods: P4-ATPase subtype expression in the cochlea was detected by immuno-fuorescence staining and quantitative real-time polymerase chain reaction (qRT-PCR) at diferent developmental stages. Hair cell specifc TMEM30A knockout mice and wild-type littermates were used for the following functional and morphological analysis. Auditory function was evaluated by auditory brainstem response. We investigated hair cell and stereocilia morphological changes by immunofuorescence staining. Scanning electron microscopy was applied to observe the stereocilia ultrastructure. Diferentially expressed transcriptomes were analyzed based on RNA-sequencing data from knock-out and wild-type mouse cochleae. Diferentially expressed genes were verifed by qRT-PCR.
Results: TMEM30A and subtypes of P4-ATPase are expressed in the mouse cochlea in a temporal-dependent pattern. Deletion of TMEM30A in hair cells impaired hearing onset due to progressive hair cell loss. The disrupted kinocilia placement and irregular distribution of spectrin-α in cuticular plate indicated the hair cell planar polarity disruption in TMEM30A deletion hair cells. Hair cell degeneration begins at P7 and finishes around P14. Transcriptional analysis indicates that the focal adhesion pathway and stereocilium tip-related genes changed dramatically. Without the TMEM30A chaperone, excessive ATP8A2 accumulated in the cytoplasm, leading to overwhelming endoplasmic reticulum stress, which eventually contributed to hair cell death.
Conclusions: Deletion of TMEM30A led to disrupted planar polarity and stereocilia bundles, and finally led to hair cell loss and auditory dysfunction. TMEM30A is essential for hair cell polarity maintenance and membrane homeostasis. Our study highlights a pivotal role of TMEM30A in the postnatal development of hair cells and reveals the possible mechanisms underlying P4-ATPase-related genetic hearing loss.
Keywords P4-ATPase, Flippases, Planar cell polarity, ER stress, Hearing loss
Address and Contact Information 1 Department of Otolaryngology Head & Neck Surgery, Shanghai Sixth People’s Hospital Afliated to Shanghai Jiao Tong University School of Medicine, 1301 Research Bldg, 600 Yishan Rd, Shanghai, China
2 Otolaryngology Institute of Shanghai Jiao Tong University, 600 Yishan Rd, Shanghai 200233, China
3 Health Management Center, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
*Corresponding author: gyang321@sjtu.edu.cn
Yazhi Xing and Kun Peng contributed equally to this work
Read full article at BMC

No.  24DOI: 10.1186/s11658-023-00435-y Volume 28 (2023) - 28:24
Title EXTRACELLULAR VESICLES DERIVED FROM CD4+ T CELLS CARRY DGKK TO PROMOTE SEPSIS-INDUCED LUNG INJURY BY REGULATING OXIDATIVE STRESS AND INFAMMATION
Authors Guo‐wei Tu1†, Yi Zhang2†, Jie‐fei Ma3†, Jun‐yi Hou1, Guang‐wei Hao, Ying Su1, Jing‐chao Luo1, Lulu Sheng4* and Zhe Luo1,3,5,6*
Abstract Background: Sepsis is an abnormal immune response after infection, wherein the lung is the most susceptible organ to fail, leading to acute lung injury. To overcome the limitations of current therapeutic strategies and develop more specifc treatment, the infammatory process, in which T cell-derived extracellular vesicles (EVs) play a central role, should be explored deeply.
Methods: Liquid chromatography–tandem mass spectrometry was performed for serum EV protein profling. The serum diacylglycerol kinase kappa (DGKK) and endotoxin contents of patients with sepsis-induced lung injury were measured. Apoptosis, oxidative stress, and infammation in A549 cells, bronchoalveolar lavage fuid, and lung tissues of mice were measured by flow cytometry, biochemical analysis, enzyme-linked immunosorbent assay, quantitative real-time polymerase chain reaction, and western blot.
Results: DGKK, the key regulator of the diacylglycerol (DAG)/protein kinase C (PKC) pathway, exhibited elevated expression in serum EVs of patients with sepsis-induced lung injury and showed strong correlation with sepsis severity and disease progression. DGKK was expressed in CD4+ T cells under regulation of the NF-κB pathway and delivered by EVs to target cells, including alveolar epithelial cells. EVs produced by CD4+ T lymphocytes exerted toxic efects on A549 cells to induce apoptotic cell death, oxidative cell damage, and infammation. In mice with sepsis induced by cecal ligation and puncture, EVs derived from CD4+ T cells also promoted tissue damage, oxidative stress, and infammation in the lungs. These toxic efects of T cell-derived EVs were attenuated by the inhibition of PKC and NOX4, the downstream efectors of DGKK and DAG.
Conclusions: This approach established the mechanism that T-cell-derived EVs carrying DGKK triggered alveolar epithelial cell apoptosis, oxidative stress, infammation, and tissue damage in sepsis-induced lung injury through the DAG/PKC/NOX4 pathway. Thus, T-cell-derived EVs and the elevated distribution of DGKK should be further investigated to develop therapeutic strategies for sepsis-induced lung injury.
Keywords Sepsis-induced lung injury, DGKK, Extracellular vesicles, Oxidative stress, Infammation
Address and Contact Information 1 Cardiac Intensive Care Center, Zhongshan Hospital, Fudan University, Shanghai, China
2 Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
3 Department of Critical Care Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
4 Department of Emergency Medicine, Shanghai Jiao Tong University Afliated Sixth People’s Hospital, Shanghai, China
5 Shanghai Key Laboratory of Lung Infammation and Injury, Shanghai, China
6 Department of Critical Care Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Shanghai, China
*Corresponding author: yl20111211856@sjtu.edu.cn; luo.zhe@zs-hospital.sh.cn
Guo-wei Tu, Yi Zhang, and Jie-fei Ma contributed equally to this article and are co-frst authors.
Read full article at BMC

No.  25DOI: 10.1186/s11658-023-00434-z Volume 28 (2023) - 28:25
Title p20BAP31 INDUCES CELL APOPTOSISVIA BOTH AIF CASPASE-INDEPENDENT AND THE ROS/JNK MITOCHONDRIAL PATHWAY IN COLORECTAL CANCER
Authors Xiaohan Jiang, Guoxun Li, Benzhi Zhu, Jingnan Zang, Tian Lan, Rui Jiang* and Bing Wang*
Abstract Background: During cell apoptosis, the C-terminus of BAP31 is cleaved by caspase-8 and generates p20BAP31, which has been shown to induce an apoptotic pathway between the endoplasmic reticulum (ER) and mitochondria. However, the underlying mechanisms of p20BAP31 in cell apoptosis remains unclear.
Methods: We compared the efects of p20BAP31 on cell apoptosis in six cell lines and selected the most sensitive cells. Functional experiments were conducted, including Cell Counting Kit 8 (CCK-8), reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) assay. Then, cell cycle and apoptosis were investigated by flow cytometry and verifed by immunoblotting. Next, NOX inhibitors (ML171 and apocynin), ROS scavenger (NAC), JNK inhibitor (SP600125), and caspase inhibitor (Z-VAD-FMK) were used to further investigate the underlying mechanisms of p20BAP31 on cell apoptosis. Finally, apoptosis-inducing factor (AIF) translocation from the mitochondria to the nuclei was verifed by immunoblotting and immunofuorescence assay.
Results: We found that overexpression of p20BAP31 indeed induced apoptosis and had a much greater sensitivity in HCT116 cells. Furthermore, the overexpression of p20BAP31 inhibited cell proliferation by causing S phase arrest. Further study revealed that p20BAP31 reduced MMP, with a signifcant increase in ROS levels, accompanied by the activation of the MAPK signaling pathway. Importantly, the mechanistic investigation indicated that p20BAP31 induces mitochondrial-dependent apoptosis by activating the ROS/JNK signaling pathway and induces caspase-independent apoptosis by promoting the nuclear translocation of AIF.
Conclusions: p20BAP31 induced cell apoptosis via both the ROS/JNK mitochondrial pathway and AIF caspase-independent pathway. Compared with antitumor drugs that are susceptible to drug resistance, p20BAP31 has unique advantages for tumor therapy.
Keywords p20BAP31, Apoptosis, AIF, ROS/JNK pathway, Colorectal cancer
Address and Contact Information College of Life and Health Science, Northeastern University, 195 Chuangxin Road, Hunnan District, Shenyang, Liaoning Province, China
*Corresponding author: jiangrui@mail.neu.edu.cn; wangbing@mail.neu.edu.cn
Read full article at BMC

No.  26DOI: 10.1186/s11658-023-00440-1 Volume 28 (2023) - 28:26
Title rt269L-TYPE HEPATITIS B VIRUS (HBV) IN GENOTYPE C INFECTION LEADS TO IMPROVED MITOCHONDRIAL DYNAMICS VIA THE PERK–eIF2α–ATF4 AXIS IN AN HBX PROTEIN-DEPENDENT MANNER
Authors Yu‐Min Choi1, Dong Hyun Kim1, Junghwa Jang1, Won Hyeok Choe2 and Bum‐Joon Kim1,3,4,5,6*
Abstract Background: In our previous report, the rt269I type versus the rt269L type in genotype C2 infection led to poor clinical outcomes and enhanced mitochondrial stress in infected hepatocytes. Here, we sought to investigate diferences between the rt269L and rt269I types in mitochondrial functionality in hepatitis B virus (HBV) genotype C2 infection, mainly focusing on endoplasmic reticulum (ER) stress-mediated autophagy induction as an upstream signal.
Methods: Mitochondrial functionality, ER stress signaling, autophagy induction, and apoptotic cell death between rt269L-type and rt269I-type groups were investigated via in vitro and in vivo experiments. Serum samples were collected from 187 chronic hepatitis patients who visited Konkuk or Seoul National University Hospital.
Results: Our data revealed that genotype C rt269L versus rt269I infection led to improved mitochondrial dynamics and enhanced autophagic fux, mainly due to the activation of the PERK–eIF2α–ATF4 axis. Furthermore, we demonstrated that the traits found in genotype C rt269L infection were mainly due to increased stability of the HBx protein after deubiquitination. In addition, clinical data using patient sera from two independent Korean cohorts showed that, compared with rt269I, rt269L in infection led to lower 8-OHdG levels, further supporting its improved mitochondrial quality control.
Conclusion: Our data showed that, compared with the rt269I type, the rt269L type, which presented exclusively in HBV genotype C infection, leads to improved mitochondrial dynamics or bioenergetics, mainly due to autophagy induction via activation of the PERK–eIF2α–ATF4 axis in an HBx protein-dependent manner. This suggests that HBx stability and cellular quality control in the rt269L type predominating in genotype C endemic areas could at least partly contribute to some distinctive traits of genotype C infection, such as higher infectivity or longer duration of the hepatitis B e antigen (HBeAg) positive stage.
Keywords Mitochondrial functionality, ER stress, Autophagy, HBx stability, Deubiquitination
Address and Contact Information 1 Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul 110‐799, Republic of Korea
2 Department of Internal Medicine, Konkuk University School of Medicine, Seoul 05030, Republic of Korea
3 Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
4 Liver Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea
5 Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea
6 Seoul National University Medical Research Center (SNUMRC), Seoul 03080, Korea
*Corresponding author: kbumjoon@snu.ac.kr
Read full article at BMC

No.  27DOI: 10.1186/s11658-023-00439-8 Volume 28 (2023) - 28:27
Title IFN-γ BLOCKADE AFTER GENETIC INHIBITION OF PD-1 AGGRAVATES SKELETAL MUSCLE DAMAGE AND IMPAIRS SKELETAL MUSCLE REGENERATION
Authors Shuzhao Zhuang1,2,3, Aaron Russell3, Yifan Guo1,2, Yingying Xu1,2 and Weihua Xiao1,2*
Abstract Background: Innate immune responses play essential roles in skeletal muscle recovery after injury. Programmed cell death protein 1 (PD-1) contributes to skeletal muscle regeneration by promoting macrophage proinfammatory to anti-infammatory phenotype transition. Interferon (IFN)-γ induces proinfammatory macrophages that appear to hinder myogenesis in vitro. Therefore, we tested the hypothesis that blocking IFN-γ in PD-1 knockout mice may dampen infammation and promote skeletal muscle regeneration via regulating the macrophage phenotype and neutrophils.
Methods: Anti-IFN-γ antibody was administered in PD-1 knockout mice, and cardiotoxin (CTX) injection was performed to induce acute skeletal muscle injury. Hematoxylin and eosin (HE) staining was used to view morphological changes of injured and regenerated skeletal muscle. Masson’s trichrome staining was used to assess the degree of fbrosis. Gene expressions of proinfammatory and anti-infammatory factors, fibrosis-related factors, and myogenic regulator factors were determined by real-time polymerase chain reaction (PCR). Changes in macrophage phenotype were examined by western blot and real-time PCR. Immunofuorescence was used to detect the accumulation of proinfammatory macrophages, anti-infammatory macrophages, and neutrophils.
Results: IFN-γ blockade in PD-1 knockout mice did not alleviate skeletal muscle damage or improve regeneration following acute cardiotoxin-induced injury. Instead, it exacerbated skeletal muscle infammation and fibrosis, and impaired regeneration via inhibiting macrophage accumulation, blocking macrophage proinfammatory to anti-infammatory transition, and enhancing infltration of neutrophils.
Conclusion: IFN-γ is crucial for efcient skeletal muscle regeneration in the absence of PD-1.
Keywords Skeletal muscle, Macrophage, Neutrophil, PD-1, IFN-γ
Address and Contact Information 1 Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
2 Key Laboratory of Exercise and Health Sciences, Shanghai University of Sport, Ministry of Education, Shanghai, China
3 Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
*Corresponding author: xiao_weihua@163.com
Read full article at BMC

No.  28DOI: 10.1186/s11658-023-00442-z Volume 28 (2023) - 28:28
Title BLOOD-BASED LIQUID BIOPSY: INSIGHTS INTO EARLY DETECTION, PREDICTION, AND TREATMENT MONITORING OF BLADDER CANCER
Authors Shijie Li1†, Kerong Xin1†, Shen Pan2†, Yang Wang3†, Jianyi Zheng1, Zeyu Li1, Xuefeng Liu1, Bitian Liu1*, Zhenqun Xu1* and Xiaonan Chen1*
Abstract Bladder cancer (BC) is a clinical challenge worldwide with late clinical presentation, poor prognosis, and low survival rates. Traditional cystoscopy and tissue biopsy are routine methods for the diagnosis, prognosis, and monitoring of BC. However, due to the heterogeneity and limitations of tumors, such as aggressiveness, high cost, and limited applicability of longitudinal surveillance, the identifcation of tumor markers has attracted signifcant attention in BC. Over the past decade, liquid biopsies (e.g., blood) have proven to be highly efcient methods for the discovery of BC biomarkers. This noninvasive sampling method is used to analyze unique tumor components released into the peripheral circulation and allows serial sampling and longitudinal monitoring of tumor progression. Several liquid biopsy biomarkers are being extensively studied and have shown promising results in clinical applications of BC, including early detection, detection of microscopic residual disease, prediction of recurrence, and response to therapy. Therefore, in this review, we aim to provide an update on various novel blood-based liquid biopsy markers and review the advantages and current limitations of liquid biopsy in BC therapy. The role of blood-based circulating tumor cells, circulating tumor DNA, cell-free RNA, exosomes, metabolomics, and proteomics in diagnosis, prognosis, and treatment monitoring, and their applicability to the personalized management of BC, are highlighted.
Keywords Bladder cancer, Liquid biopsy, Circulating tumor cells, Circulating tumor DNA, Cell-free RNA, Exosomes, Metabolomics, Proteomics, Clinical application
Address and Contact Information 1 Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, People’s Republic of China
2 Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, People’s Republic of China
3 Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning, People’s Republic of China
*Corresponding author: liu_bitian@163.com; zqxu@cmu.edu.cn; chenxn@cmu.edu.cn Shijie Li, Kerong Xin, Shen Pan, and Yang Wang contributed equally to this work.
Read full article at BMC

No.  29DOI: 10.1186/s11658-023-00444-x Volume 28 (2023) - 28:29
Title Correction: Inhibition of PAD4 enhances radiosensitivity and inhibits aggressive phenotypes of nasopharyngeal carcinoma cells
Authors Hao Chen1†, Min Luo1†, Xiangping Wang1, Ting Liang1, Chaoyuan Huang1, Changjie Huang1* and Lining Wei2*
Abstract Correction: Cellular & Molecular Biology Letters 2021, 26(1):9
https://doi.org/10.1186/s11658-021-00251-2

Following publication of the original article [1], the authors informed us that the images of Transwell assays (Figs. 2G, 3F, 4F) were incorrect. Te correct images are given below. Te replacement of images does not afect the original conclusion.
Keywords
Address and Contact Information 1 Department of Oncology, The Second Nanning People’s Hospital, No. 13 Dancun Road, Jiangnan District, Nanning 530031, Guangxi, China
2 Department of Endoscopy, The Afliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
*Corresponding author: hcj1215423068@hotmail.Com; weilininggudy@163.com
Hao Chen and Min Luo contributed equally to this work
The original article can be found online at https://doi.org/10.1186/s11658-021-00251-2.
Read full article at BMC

No.  30DOI: 10.1186/s11658-023-00441-0 Volume 28 (2023) - 28:30
Title CAN PRECANCEROUS STEM CELLS BE RISK MARKERS FOR MALIGNANT TRANSFORMATION IN THE ORAL MUCOSA?
Authors Shan Wang1,2*, Liu Ying3, Shu‐Yi Yu4, Jie Bai5* and Chunbo Hao6*
Abstract Accurate assessment of the carcinogenic potential of oral mucosal diseases can signifcantly reduce the prevalence of oral cancer. We speculate that precancerous stem cells (pCSCs) arise during the evolution of carcinomas based on long-term experimental findings, published literature, and the cancer stem cell (CSC) theory, wherein pCSCs exist in precancerous lesions and have characteristics of both CSCs and normal stem cells. This apparently contradictory feature may be the foundation of the reversible transformation of precancerous lesions. Predicting malignant transformation in potentially malignant oral illnesses would allow for focused treatment, prognosis, and secondary prevention. Currently available clinical assays for chromosomal instability and DNA aneuploidy have several defciencies. We hope that our study will increase attention to pCSC research and lead to the development of novel strategies for the prevention and treatment of oral cancer by identifying pCSC markers.
Keywords Precancerous stem cells, Cancer stem cells, Oral cancer, Dysplasia, Malignant transformation
Address and Contact Information 1 Department of Oral Pathology, School of Stomatology, Hainan Medical University, Haikou 571199, People’s Republic of China
2 Department of Stomatology, The Second Afliated Hospital of Hainan Medical University, Haikou 570216, People’s Republic of China
3 College of Pharmacy, Hainan Medical University, Haikou 571199, People’s Republic of China
4 Pharmacy Department, First Afliated Hospital of Jiamusi University, Jiamusi 154003, People’s Republic of China
5 Department of Ophthalmology, the Fourth Afliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, People’s Republic of China
6 Department of Stomatology, Hainan General Hospital (Hainan Afliated Hospital of Hainan Medical University), Haikou 570100, People’s Republic of China
*Corresponding author: birchtree20032003@126.com; hy0210032@hainanmc.edu.cn; 8020234@zju.edu.cn; haochunbonuli@sina.com
Read full article at BMC

No.  31DOI: 10.1186/s11658-023-00443-y Volume 28 (2023) - 28:31
Title RPL21 INTERACTS WITH LAMP3 TO PROMOTE COLORECTAL CANCER INVASION AND METASTASIS BY REGULATING FOCAL ADHESION FORMATION
Authors Jiaxian Zhu1,3, Ting Long1, Lingfang Gao1, Yan Zhong1, Ping Wang1,3, Xiaoyan Wang2,3,4, Zuguo Li1,4* and Zhiyan Hu2,3,4*
Abstract Background: Metastasis is the leading cause of death among patients with colorectal cancer (CRC). Therefore, it is important to explore the molecular mechanisms of metastasis to develop efective therapeutic targets for CRC. In the present study, ribosomal protein L21 (RPL21) was considered as being involved in promoting CRC metastasis, yet the underlying mechanism requires further investigation.
Methods: Immunohistochemistry, western blotting, and quantitative reverse transcription polymerase chain reaction were performed to measure the expression of RPL21 and lysosome-associated membrane protein 3 (LAMP3) in CRC tissues and cells. Wound healing, transwell migration, and invasion assays were performed to study the migration and invasion of cultured CRC cells. An orthotopic CRC mouse model was developed to investigate the metastatic ability of CRC. Transcriptome sequencing was conducted to identify the genes related to RPL21. The dual-luciferase reporter gene assay was performed to determine the transcriptional activity of transcription factor EB (TFEB). The GST/His pull-down assay was performed to investigate the specifc binding sites of RPL21 and LAMP3. The cell adhesion assay was performed to determine the adhesion ability of CRC cells. Immunofuorescence staining was performed to observe focal adhesions (FAs).
Results: RPL21 was highly expressed in CRC, contributing to tumor invasiveness and poor patient prognosis. Functionally, RPL21 promoted the migration and invasion of CRC cells in vitro and tumor metastasis in vivo. Moreover, LAMP3 was identifed as being highly related to RPL21 and was essential in promoting the migration and invasion of CRC cells. Mechanistically, RPL21 activated the transcriptional function of TFEB to upregulate LAMP3 expression. RPL21 directly bound to the aa 341–416 domain of LAMP3 via its aa 1–40 and aa 111–160 segments. The combination of RPL21 and LAMP3 enhanced the stability of the RPL21 protein by suppressing the degradation of the ubiquitin–proteasome system. Furthermore, RPL21 and LAMP3 promoted the formation of immature FAs by activating the FAK/paxillin/ERK signaling pathway.
Conclusions: RPL21 promoted invasion and metastasis by regulating FA formation in a LAMP3-dependent manner during CRC progression. The interaction between RPL21 and LAMP3 may function as a potential therapeutic target against CRC.
Keywords RPL21, LAMP3, TFEB, Focal adhesion, Colorectal cancer, Migration, Invasion, Metastasis
Address and Contact Information 1 Department of Pathology, Shenzhen Hospital, Southern Medical University, 1333 Xinhu Road, Shenzhen 518101, Guangdong, People’s Republic of China
2 Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, Guangdong, People’s Republic of China
3 Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, People’s Republic of China
4 Key Laboratory of Molecular Tumour Pathology of Guangdong Province, Guangzhou 510515, Guangdong, People’s Republic of China
*Corresponding author: lizg@smu.edu.cn; zhenyucai2011@163.com
Read full article at BMC

No.  32DOI: 10.1186/s11658-023-00445-w Volume 28 (2023) - 28:32
Title KIAA1429-MEDIATED m6A MODIFCATION OF CHST11 PROMOTES PROGRESSION OF DIFUSE LARGE B-CELL LYMPHOMA BY REGULATING Hippo–YAP PATHWAY
Authors Xiaomin Chen1, Tiange Lu1, Yiqing Cai1, Yang Han2, Mengfei Ding1, Yurou Chu1, Xiangxiang Zhou1,2,3,4,5* and Xin Wang1,2,3,4,5*
Abstract Background: N6-methyladenosine (m6A) has been shown to participate in various essential biological processes by regulating the level of target genes. However, the function of m6A modifcation mediated by KIAA1429 [alias virus-like m6A methyl-transferase-associated protein (VIRMA)] during the progression of difuse large B-cell lymphoma (DLBCL) remains undefned.
Methods: The expression and clinical signifcance of KIAA1429 were verifed by our clinical data. CRISPR/Cas9 mediated KIAA1429 deletion, and CRISPR/dCas9-VP64 for activating endogenous KIAA1429 was used to evaluate its biological function. RNA sequencing (RNA-seq), methylated RNA immunoprecipitation sequencing (MeRIP-seq), RNA immunoprecipitation (RIP) assays, luciferase activity assay, RNA stability experiments, and co-immunoprecipitation were performed to investigate the regulatory mechanism of KIAA1429 in DLBCL. Tumor xenograft models were established for in vivo experiments.
Results: Dysregulated expression of m6A regulators was observed, and a novel predictive model based on m6A score was established in DLBCL. Additionally, elevated KIAA1429 expression was associated with poor prognosis of patients with DLBCL. Knockout of KIAA1429 repressed DLBCL cell proliferation, facilitated cell cycle arrest in the G2/M phase, induced apoptosis in vitro, and inhibited tumor growth in vivo. Furthermore, carbohydrate sulfotransferase 11 (CHST11) was identifed as a downstream target of KIAA1429, which mediated m6A modifcation of CHST11 mRNA and then recruited YTHDF2 for reducing CHST11 stability and expression. Inhibition of CHST11 diminished MOB1B expression, resulting in inactivation of Hippo–YAP signaling, reprogramming the expression of Hippo target genes.
Conclusions: Our results revealed a new mechanism by which the Hippo–YAP pathway in DLBCL is inactivated by KIAA1429/YTHDF2-coupled epitranscriptional repression of CHST11, highlighting the potential of KIAA1429 as a novel predictive biomarker and therapeutic target for DLBCL progression.
Keywords Difuse large B-cell lymphoma, N6-methyladenosine, KIAA1429, YTHDF2, CHST11, Hippo–YAP
Address and Contact Information 1 Department of Hematology, Shandong Provincial Hospital, Shandong University, No.324, Jingwu Road, Jinan 250021, Shandong, China
2 Department of Hematology, Shandong Provincial Hospital Afliated to Shandong First Medical University, Jinan 250021, Shandong, China
3 4 Branch of National Clinical Research Center for Hematologic Diseases, Jinan 250021, Shandong, China
5 National Clinical Research Center for Hematologic Diseases, The First Afliated Hospital of Soochow University, Suzhou 251006, China
*Corresponding author: xiangxiangzhou@sdu.edu.cn; xinw007@126.com
Read full article at BMC

No.  33DOI: 10.1186/s11658-023-00438-9 Volume 28 (2023) - 28:33
Title DECIPHERING STAT3 SIGNALING POTENTIAL IN HEPATOCELLULAR CARCINOMA: TUMORIGENESIS, TREATMENT RESISTANCE, AND PHARMACOLOGICAL SIGNIFCANCE
Authors Mehrdad Hashemi1,2, Eisa Sabouni3, Parham Rahmanian3, Maliheh Entezari1,2, Mahsa Mojtabavi4, Behnaz Raei1, Mohammad Arad Zandieh5, Mitra Behroozaghdam1, Sepideh Mirzaei6, Kiavash Hushmandi5, Noushin Nabavi7, Shokooh Salimimoghadam8, Jun Ren9, Mohsen Rashidi10,11*, Rasoul Raesi12,13*, Afshin Taheriazam1,14*, Athanasios Alexiou15,16, Marios Papadakis17* and Shing Cheng Tan18
Abstract Hepatocellular carcinoma (HCC) is considered one of the greatest challenges to human life and is the most common form of liver cancer. Treatment of HCC depends on chemotherapy, radiotherapy, surgery, and immunotherapy, all of which have their own drawbacks, and patients may develop resistance to these therapies due to the aggressive behavior of HCC cells. New and efective therapies for HCC can be developed by targeting molecular signaling pathways. The expression of signal transducer and activator of transcription 3 (STAT3) in human cancer cells changes, and during cancer progression, the expression tends to increase. After induction of STAT3 signaling by growth factors and cytokines, STAT3 is phosphorylated and translocated to the nucleus to regulate cancer progression. The concept of the current review revolves around the expression and phosphorylation status of STAT3 in HCC, and studies show that the expression of STAT3 is high during the progression of HCC. This review addresses the function of STAT3 as an oncogenic factor in HCC, as STAT3 is able to prevent apoptosis and thus promote the progression of HCC. Moreover, STAT3 regulates both survival- and death-inducing autophagy in HCC and promotes cancer metastasis by inducing the epithelial–mesenchymal transition (EMT). In addition, upregulation of STAT3 is associated with the occurrence of chemoresistance and radioresistance in HCC. Specifcally, non-protein-coding transcripts regulate STAT3 signaling in HCC, and their inhibition by antitumor agents may afect tumor progression. In this review, all these topics are discussed in detail to provide further insight into the role of STAT3 in tumorigenesis, treatment resistance, and pharmacological regulation of HCC.
Keywords Hepatocellular carcinoma, Liver cancer, Noncoding transcripts, STAT3, Molecular signaling
Address and Contact Information 1 Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
2 Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
3 Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran.
4 Mashhad Branch, Islamic Azad University, Mashhad, Iran.
5 Division of Epidemiology, Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
6 Department of Biology, Faculty of Science, Science and Research Branch, Islamic Azad University, Tehran, Iran.
7 Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada.
8 Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
9 Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai 200032, China.
10 Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
11 The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
12 Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran.
13 Department of Medical-Surgical Nursing, Mashhad University of Medical Sciences, Mashhad, Iran.
14 Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
15 Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia.
16 AFNP Med Austria, Vienna, Austria.
17 Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283 Wuppertal, Germany.
18 UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
*Corresponding author: dr.mohsenrashidi@yahoo.com; raesi.br881@gmail.com; a.taheriazam@iautmu.ac.ir; marios_papadakis@yahoo.gr
Read full article at BMC

No.  34DOI: 10.1186/s11658-023-00448-7 Volume 28 (2023) - 28:34
Title CircRNF10-DHX15 INTERACTION SUPPRESSED BREAST CANCER PROGRESSION BY ANTAGONIZING DHX15-NF-κB p65 POSITIVE FEEDBACK LOOP
Authors Wenfang Zheng1, Xuehui Wang1, Yunhe Yu1, Changle Ji1 and Lin Fang1*
Abstract Background: Breast cancer (BC) is a common threat to women. The continuous activation of nuclear factor kappa B (NF-κB) signaling pathway contributes to the development of BC. This study aimed to investigate the role of a circular RNA (circRNF10) in BC progression and regulating NF-κB signaling pathway.
Methods: Bioinformatics analysis, RT-qPCR, subcellular fractionation, FISH, RNase R treatment, and actinomycin D assay were used to explore the expression and characteristics of circRNF10 in BC. The biological functions of circRNF10 in BC were analyzed by MTT assay, colony formation assay, wound healing assay, and Transwell assay. RNA pulldown and RIP assay were used to identify the interaction between circRNF10 and DEAH (Asp-Glu-Ala-His) box helicase 15 (DHX15). The impact of circRNF10-DHX15 interaction on NF-κB signaling pathway was explored by western blot, IF, and co-IP. Furthermore, dual-luciferase reporter assay, ChIP, and EMSA were performed to assess the efect of NF-κB p65 on DHX15 transcription.
Results: CircRNF10 was downregulated in BC, and lower expression of circRNF10 was related to poor prognosis of patients with BC. CircRNF10 inhibited the proliferation and migration of BC. Mechanically, circRNF10-DHX15 interaction sequestered DHX15 from NF-κB p65, thereby inhibiting the activation of NF-κB signaling pathway. On the other hand, NF-κB p65 enhanced DHX15 transcription by binding to the promoter of DHX15. Altogether, circRNF10 impaired the DHX15-NF-κB p65 positive feedback loop and suppressed the progression of BC.
Conclusion: CircRNF10-DHX15 interaction suppressed the DHX15-NF-κB p65 positive feedback loop, thereby inhibiting BC progression. These findings provide new insights in the continuous activation of NF-κB signaling pathway and raised potential therapeutic approach for BC treatment.
Keywords circRNF10, DHX15, p65, Feedback, Breast cancer
Address and Contact Information 1 Department of Breast and Thyroid Surgery, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, 301 Yanchangzhong Road, Shanghai 200072, People’s Republic of China
*Corresponding author: 1200038@tongji.edu.cn
Read full article at BMC

No.  35DOI: 10.1186/s11658-022-00415-8 Volume 28 (2023) - 28:35
Title PROTECTION OF MELATONIN TREATMENT AND COMBINATION WITH TRADITIONAL ANTIBIOTICS AGAINST SEPTIC MYOCARDIAL INJURY
Authors Wencheng Di1†, Zhenxiao Jin2†, Wangrui Lei3,4†, Qiong Liu3,4, Wenwen Yang3,4, Shaofei Zhang3,4, Chenxi Lu3,4, Xiaoling Xu3,4* and Huadong Zhao5*
Abstract Background: Heart failure is a common complication of sepsis with a high mortality rate. It has been reported that melatonin can attenuate septic injury due to various properties. On the basis of previous reports, this study will further explore the efects and mechanisms of melatonin pretreatment, posttreatment, and combination with antibiotics in the treatment of sepsis and septic myocardial injury.
Methods and results: Our results showed that melatonin pretreatment showed an obvious protective efect on sepsis and septic myocardial injury, which was related to the attenuation of infammation and oxidative stress, the improvement of mitochondrial function, the regulation of endoplasmic reticulum stress (ERS), and the activation of the AMPK signaling pathway. In particular, AMPK serves as a key efector for melatonin-initiated myocardial benefts. In addition, melatonin posttreatment also had a certain degree of protection, while its efect was not as remarkable as that of pretreatment. The combination of melatonin and classical antibiotics had a slight but limited efect. RNA-seq detection clarifed the cardioprotective mechanism of melatonin.
Conclusion: Altogether, this study provides a theoretical basis for the application strategy and combination of melatonin in septic myocardial injury.
Keywords Melatonin, Septic myocardial injury, RNA-seq, Pretreatment, Posttreatment, Antibiotics
Address and Contact Information 1 Department of Cardiovascular Medicine, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, 29 Bulan Road, Shenzhen, Guangdong Province, China
2 Department of Cardiovascular Surgery, Xijing Hospital, The Airforce Military Medical University, 127 Changle West Road, Xi’an, China
3 Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, 229 Taibai North Road, Xi’an, China
4 Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi’an, China
5 Department of General Surgery, Tangdu Hospital, The Airforce Military Medical University, 1 Xinsi Road, Xi’an, China
*Corresponding author: yang200214yy@nwu.edu.cn; huadongzhaotd@163.com
Wencheng Di,Zhenxiao Jin, Wangrui Lei contributed equally as co-frst authors
Read full article at BMC

No.  36DOI: 10.1186/s11658-023-00449-6 Volume 28 (2023) - 28:36
Title MELATONIN AND ERASTIN EMERGE SYNERGISTIC ANTI-TUMOR EFECTS ON ORAL SQUAMOUS CELL CARCINOMA BY INDUCING APOPTOSIS, FERROPTOSIS, AND INHIBITING AUTOPHAGY THROUGH PROMOTING ROS
Authors Leilei Wang1, Chuan Wang2,3, Xuan Li4, Zhuoying Tao1, Wangyong Zhu5, Yuxiong Su1 and Wing Shan Choi1*
Abstract Background: Oral squamous cell carcinomas are one of the most common cancers worldwide with aggressive behavior and poor prognosis. Reactive oxygen species (ROS) are associated with cancer and cause various types of regulated cell death (RCD). Inducing the RCD pathway by modulating ROS levels is imperative to conquer cancers. The aim of this study is to investigate the synergistic anticancer efects of melatonin and erastin on ROS modulation and subsequent RCD induction.
Methods: Human tongue squamous cell carcinoma cell lines (SCC-15 cells) were treated with melatonin, erastin, or their combination. Cell viability, ROS levels, autophagy, apoptosis, and ferroptosis levels were tested according to the results of the PCR array, which were verifed with/without the induction and inhibition of ROS by H2O2 and N-acetyl-L-cysteine, respectively. In addition, a mouse-based subcutaneous oral cancer xenograft model was constructed to identify the efects of melatonin, erastin, and their combination on the autophagy, apoptosis, and ferroptosis levels in isolated tumor tissues.
Results: ROS levels were increased by the administration of melatonin at high concentrations (mM), and the combination of melatonin with erastin enhanced the levels of malonic dialdehyde, ROS, and lipid ROS, and reduced the levels of glutamate and glutathione. SQSTM1/p62, LC3A/B, cleaved caspase-3, and PARP1 protein levels in SCC-15 cells were also increased by melatoninplus erastin treatment, which further increased as ROS accumulated, and decreased as ROS levels were suppressed. Combined treatment of melatonin and erastin markedly reduced the tumor size in vivo, demonstrated no obvious systemic side efects, and signifcantly enhanced the apoptosis and ferroptosis levels in the tumor tissues, in parallel with decreased autophagy levels.
Conclusions: Melatonin combined with erastin exhibits synergistic anticancer efects without adverse reactions. Herein, this combination might become a promising alternative strategy for oral cancer treatment.
Keywords Reactive oxygen species (ROS), Oral cancer, Melatonin, Erastin, Apoptosis, Autophagy, Ferroptosis
Address and Contact Information 1 Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, 34 Hospital Road, Hong Kong SAR, China
2 The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) and Key Laboratory of Oral Biomedicine Ministry of Education (KLOBME), School and Hospital of Stomatology, Wuhan University, Wuhan, China
3 Department of Periodontology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
4 Division of Periodontology and Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
5 Department of Dental Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
*Corresponding author: drwchoi@gmail.com
Read full article at BMC

No.  37DOI: 10.1186/s11658-023-00447-8 Volume 28 (2023) - 28:37
Title THE ROLE OF NONCODING RNAs IN METABOLIC REPROGRAMMING OF CANCER CELLS
Authors Amir Saf1, Mohammadreza Saberiyan2, Mohammad‐Javad Sanaei3, Samaneh Adelian2, Fateme Davarani Asl2, Mahsa Zeinaly4, Mahdi Shamsi5 and Reza Ahmadi6*
Abstract Metabolic reprogramming is a well-known feature of cancer that allows malignant cells to alter metabolic reactions and nutrient uptake, thereby promoting tumor growth and spread. It has been discovered that noncoding RNAs (ncRNAs), including microRNA (miRNA), long noncoding RNA (lncRNA), and circular RNA (circRNA), have a role in a variety of biological functions, control physiologic and developmental processes, and even infuence disease. They have been recognized in numerous cancer types as tumor suppressors and oncogenic agents. The role of ncRNAs in the metabolic reprogramming of cancer cells has recently been noticed. We examine this subject, with an emphasis on the metabolism of glucose, lipids, and amino acids, and highlight the therapeutic use of targeting ncRNAs in cancer treatment.
Keywords Cancer, Metabolic reprogramming, Noncoding RNAs, MicroRNA, Long noncoding RNA, Circular RNA
Address and Contact Information 1 Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
2 Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
3 Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
4 Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
5 Department of Cell and Molecular Biology, Najafabad Branch, Islamic Azad University, Najafabad, Iran
6 Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Rahmatiyeh Region, Shahrekord, Iran
*Corresponding author: ahmadi.r@skums.ac.ir
Read full article at BMC

No.  38DOI: 10.1186/s11658-023-00450-z Volume 28 (2023) - 28:38
Title SINGLE‐CELL OMICS TRACES THE HETEROGENEITY OF PROSTATE CANCER CELLS AND THE TUMOR MICROENVIRONMENT
Authors Xudong Yu1,2†, Ruijia Liu1†, Wenfeng Gao1, Xuyun Wang3* and Yaosheng Zhang1,2*
Abstract Prostate cancer is one of the more heterogeneous tumour types. In recent years, with the rapid development of single-cell sequencing and spatial transcriptome technologies, researchers have gained a more intuitive and comprehensive understanding of the heterogeneity of prostate cancer. Tumour-associated epithelial cells; cancer-associated fibroblasts; the complexity of the immune microenvironment, and the heterogeneity of the spatial distribution of tumour cells and other cancer-promoting molecules play a crucial role in the growth, invasion, and metastasis of prostate cancer. Single-cell multi-omics biotechnology, especially single-cell transcriptome sequencing, reveals the expression level of single cells with higher resolution and finely dissects the molecular characteristics of diferent tumour cells. We reviewed the recent literature on prostate cancer cells, focusing on single-cell RNA sequencing. And we analysed the heterogeneity and spatial distribution diferences of diferent tumour cell types. We discussed the impact of novel single-cell omics technologies, such as rich omics exploration strategies, multi-omics joint analysis modes, and deep learning models, on future prostate cancer research. In this review, we have constructed a comprehensive catalogue of single-cell omics studies in prostate cancer. This article aimed to provide a more thorough understanding of the diagnosis and treatment of prostate cancer. We summarised and proposed several key issues and directions on applying single-cell multi-omics and spatial transcriptomics to understand the heterogeneity of prostate cancer. Finally, we discussed single-cell omics trends and future directions in prostate cancer.
Keywords Prostate cancer, Single-cell omics, Spatial transcriptomics, Heterogeneity
Address and Contact Information 1 Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
2 Beijing Tumour Minimally Invasive Medical Center of Integrated Traditional Chinese and Western Medicine, Beijing 101121, China
3 Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
*Corresponding author: xuyun1102@sina.com; zhangyaosheng@bucm.edu.cn
Xudong Yu and Ruijia Liu contributed equally to this work and share the frst authorship
Read full article at BMC

No.  39DOI: 10.1186/s11658-023-00446-9 Volume 28 (2023) - 28:39
Title OPTOGENETIC CONTROL OF YAP CAN ENHANCE THE RATE OF WOUND HEALING
Authors Pearlyn Jia Ying Toh1,2, Marius Sudol1,3 and Timothy Edward Saunders1,4,5*
Abstract Background: Tissues need to regenerate to restore function after injury. Yet, this regenerative capacity varies signifcantly between organs and between species. For example, in the heart, some species retain full regenerative capacity throughout their lifespan but human cardiac cells display a limited ability to repair the injury. After a myocardial infarction, the function of cardiomyocytes is impaired and reduces the ability of the heart to pump, causing heart failure. Therefore, there is a need to restore the function of an injured heart post myocardial infarction. We investigate in cell culture the role of the Yes-associated protein (YAP), a transcriptional co-regulator with a pivotal role in growth, in driving repair after injury.
Methods: We express optogenetic YAP (optoYAP) in three diferent cell lines. We characterised the behaviour and function of optoYAP using fluorescence imaging and quantitative real-time PCR of downstream YAP target genes. Mutant constructs were generated using site-directed mutagenesis. Nuclear localised optoYAP was functionally tested using wound healing assay.
Results: Utilising optoYAP, which enables precise control of pathway activation, we show that YAP induces the expression of downstream genes involved in proliferation and migration. optoYAP can increase the speed of wound healing in H9c2 cardiomyoblasts. Interestingly, this is not driven by an increase in proliferation, but by collective cell migration. We subsequently dissect specifc phosphorylation sites in YAP to identify the molecular driver of accelerated healing.
Conclusions: This study shows that optogenetic YAP is functional in H9c2 cardiomyoblasts and its controlled activation can potentially enhance wound healing in a range of conditions.
Keywords Hippo-YAP, Optogenetics, Wound healing
Address and Contact Information 1 Mechanobiology Institute, National University of Singapore, Singapore, Singapore
2 Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
3 Icahn School of Medicine at Mount Sinai, New York, NY, USA
4 Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
5 Warwick Medical School, University of Warwick, Coventry, UK
*Corresponding author: timothy.saunders@warwick.ac.uk
Read full article at BMC

No.  40DOI: 10.1186/s11658-023-00456-7 Volume 28 (2023) - 28:40
Title Galectin-1 promotes angiogenesis and chondrogenesis during antler regeneration
Authors Xunsheng Li1, Wanwan Shi1, Guanning Wei1, Jinpeng Lv1, Datao Wang1, Baorui Xing1, Jue Zhou1, Jianwei Zhao1 and Hongmei Sun1*
Abstract Background: Deer antlers are the only known mammalian structure that undergoes full regeneration. In addition, it is peculiar because when growing, it contains vascularized cartilage. The diferentiation of antler stem cells (ASCs) into chondrocytes while inducing endochondral extension of blood vessels is necessary to form antler vascularized cartilage. Therefore, antlers provide an unparalleled opportunity to investigate chondrogenesis, angiogenesis, and regenerative medicine. A study found that Galectin-1 (GAL-1), which can be used as a marker in some tumors, is highly expressed in ASCs. This intrigued us to investigate what role GAL-1 could play in antler regeneration.
Methods: We measured the expression level of GAL-1 in antler tissues and cells by immunohistochemistry, WB and QPCR. We constructed antlerogenic periosteal cells (APCs, one cell type of ASCs) with the GAL-1 gene knocked out (APCGAL-1−/−) using CRISPR-CAS9 gene editing system. The efect of GAL-1 on angiogenesis was determined by stimulating human umbilical vein endothelial cells (HUVECs) using APCGAL-1−/− conditioned medium or adding exogenous deer GAL-1 protein. The efect of APCGAL-1−/− on chondrogenic diferentiation was evaluated compared with the APCs under micro-mass culture. The gene expression pattern of APCGAL-1−/− was analyzed by transcriptome sequencing.
Results: Immunohistochemistry revealed that GAL-1 was widely expressed in the antlerogenic periosteum (AP), pedicle periosteum (PP) and antler growth center. Western blot and qRT-PCR analysis using deer cell lines further supports this result. The proliferation, migration, and tube formation assays of human umbilical vein endothelial cells (HUVECs) showed that the proangiogenic activity of APCGAL-1−/− medium was signifcantly decreased (P<0.05) compared with the APCs medium. The proangiogenic activity of deer GAL-1 protein was further confrmed by adding exogenous deer GAL-1 protein (P<0.05). The chondrogenic diferentiation ability of APCGAL-1−/− was impeded under micro-mass culture. The terms of GO and KEGG enrichment of the diferentially expressed genes (DEGs) of APCGAL-1−/− showed that down-regulated expression of pathways associated with deer antler angiogenesis, osteogenesis and stem cell pluripotency, such as the PI3K-AKT signaling pathway, signaling pathways regulating pluripotency of stem cells and TGF-β signaling pathway.
Conclusions: Deer GAL-1, has strong angiogenic activity, is widely and highly expressed in deer antler. The APCs can induce angiogenesis by secreting GAL-1. The knockout of GAL-1 gene of APCs damaged its ability to induce angiogenesis and diferentiate into chondrocytes. This ability is crucial to the formation of deer antler vascularized cartilage. Moreover, Deer antlers ofer a unique model to explore explore how angiogenesis at high levels of GAL-1 expression can be elegantly regulated without becoming cancerous.
Keywords Galectin-1, Antler stem cells, Regeneration, Crispr/cas9, Angiogenesis, Chondrogenesis
Address and Contact Information 1 Institute of Special Economic Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Jilin, China
*Corresponding author: Sunhongmei@caas.cn
Read full article at BMC

No.  41DOI: 10.1186/s11658-023-00454-9 Volume 28 (2023) - 28:41
Title PROFLING THE IMPACT OF THE PROMOTERS ON CRISPR-Cas12a SYSTEM IN HUMAN CELLS
Authors Jinhe Li2†, Qinchun Liang2,3†, HuaPing Zhou1, Ming Zhou1* and Hongxin Huang1*
Abstract The plasmid vector platform is the most commonly used vector for the expression of the versatile CRISPR-Cas technique and the promoter is a crucial element for the expression vector, thus profling the impact of the promoters on CRISPR editors provides the basic information for the gene-editing toolkits and can be a guideline for its design. Herein, we made a parallel comparison among four commonly used promoters (CAG,~1700 bp; EF1a core,~210 bp; CMV, ~500 bp; and PGK,~500 bp) in CRISPR-Cas12a system in mammalian cells to explore the impact of promoters on this powerful tool. We found that without badly damaging targeting specifcity, the CAG promoter-driving Cas12a editor exhibited the most active (eficiency takes as 100%, specificity index= ~75%) in genomic cleavage, multiplex editing, transcriptional activation, and base editing, followed by promoter CMV (eficiency=70~90% (vs CAG), specificity index= ~78%), and then EF1a core and PGK (both efciency=40–60%, vs CAG) but with higher specificity (specificity index= ~84% and~82%, respectively). Therefore, CAG is recommended in the CRISPR-Cas12a system for the applications that need a robust editing activity but without size limitation, CMV mostly can be an alternative for CAG when requiring a smaller space, EF1a is similar to PGK with relatively high specificity, but has a smaller size, thus is more suitable for in vivo therapeutic applications. The data outlined the properties of the widely used promoters in the CRISPR-Cas12a system, which can be a guide for its applications and can be a useful resource for the gene-editing feld.
Keywords CRISPR-Cas system, Promoters, Impact, Comparison, Plasmid design
Address and Contact Information 1 Afliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
2 The Third Afliated Hospital of Southern Medical University, Guangzhou 510630, China
3 Guangzhou Key Laboratory of Neuropathic Pain Mechanism at Spinal Cord Level, Guangzhou 510630, China
*Corresponding author: zhouming@gzhmu.edu.cn; huanghx08@gzhmu.edu.cn
Jinhe Li and Qinchun Liang equally contributed to this work.
Read full article at BMC

No.  42DOI: 10.1186/s11658-023-00457-6 Volume 28 (2023) - 28:42
Title EMPAGLIFOZIN IMPROVES RENAL ISCHEMIA–REPERFUSION INJURY BY REDUCING INFAMMATION AND ENHANCING MITOCHONDRIAL FUSION THROUGH AMPK–OPA1 PATHWAY PROMOTION
Authors Wenbo Yang1†, Xiaoli Li2†, Liujie He3†, Shuyang Zhu3†, Shicong Lai1, Xiaopeng Zhang1, Zixiong Huang1, Biyue Yu4, Chunping Cui5 and Qiang Wang1*
Abstract Background: Renal ischemia–reperfusion injury (IRI) is one reason for renal transplantation failure. Recent studies have shown that mitochondrial dynamics is closely related to IRI, and that inhibition or reversal of mitochondrial division protects organs against IRI. Optic atrophy protein 1 (OPA1), an important factor in mitochondrial fusion, has been shown to be upregulated by sodium-glucose cotransporter 2 inhibitor (SGLT2i). Also, the antiinflammatory efects of SGLT2i have been demonstrated in renal cells. Thus, we hypothesized that empagliflozin could prevent IRI through inhibiting mitochondrial division and reducing infammation.
Methods: Using hematoxylin–eosin staining, enzyme linked immunosorbent assay (ELISA), flow cytometry, immunofuorescent staining, terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) staining, real-time PCR, RNA-sequencing, and western blot, we analyzed renal tubular tissue from in vivo and in vitro experiments.
Results: Through animal experiments and sequencing analysis, we first confirmed the protection against IRI and the regulation of mitochondrial dynamics-related factors and infammatory factors by empagliflozin pretreatment. Then, through hypoxia/reoxygenation (H/R) cellular experiments, we confirmed that empagliflozin could inhibit mitochondrial shortening and division and upregulate OPA1 in human renal tubular epithelial cell line (HK-2) cells. Subsequently, we knocked down OPA1, and mitochondrial division and shortening were observed, which could be alleviated by empagliflozin treatment. Combined with the previous results, we concluded that OPA1 downregulation leads to mitochondrial division and shortening, and empagliflozin can alleviate the condition by upregulating OPA1. We further explored the pathway through which empagliflozin functions. Related studies have shown the activation of AMPK pathway by empagliflozin and the close correlation between the AMPK pathway and OPA1. In our study, we blocked the AMPK pathway, and OPA1 upregulation by empagliflozin was not observed, thus demonstrating the dependence of empagliflozin on the AMPK pathway.
Conclusion: The results indicated that empagliflozin could prevent or alleviate renal IRI through antiinfammatory efects and the AMPK–OPA1 pathway. Ischemia–reperfu-sion injury is an inevitable challenge in organ transplantation. It is necessary to develop a new therapeutic strategy for IRI prevention in addition to refning the transplantation process. In this study, we confirmed the preventive and protective efects of empagliflozin in renal ischemia–reperfusion injury. Based on these findings, empagliflozin is promising to be a preventive agent for renal ischemia–reperfusion injury and can be applied for preemptive administration in kidney transplantation.
Keywords Renal ischemia–reperfusion injury, Empaglifozin, SGLT2i, Infammation, Mitochondrial dynamics, OPA1, AMPK signaling
Address and Contact Information 1 Department of Urology, Peking University People’s Hospital, Beijing 100044, China
2 Department of the Eighth Healthcare, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing 100853, People’s Republic of China
3 Naval Medical University, Shanghai 200433, China
4 School of Life Sciences, Hebei University, Baoding 071002, Hebei, China
5 State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China
*Corresponding author: wq301135@126.com
Wenbo Yang, Xiaoli Li, Liujie He and Shuyang Zhu contributed equally to this article.
Read full article at BMC

No.  43DOI: 10.1186/s11658-023-00452-x Volume 28 (2023) - 28:43
Title COMPARISON BETWEEN 5 EXTRACTIONS METHODS IN EITHER PLASMA OR SERUM TO DETERMINE THE OPTIMAL EXTRACTION AND MATRIX COMBINATION FOR HUMAN METABOLOMICS
Authors Maryne Lepoittevin1, Quentin Blancart‐Remaury2, Thomas Kerforne1,3, Luc Pellerin1,4, Thierry Hauet1,4,5 and Raphael Thuillier1,4,5*
Abstract Background: Although metabolomics continues to expand in many domains of research, methodological issues such as sample type, extraction and analytical protocols have not been standardized, impeding proper comparison between studies and future research.
Methods: In the present study, five solvent-based and solid-phase extraction methods were investigated in both plasma and serum. All these extracts were analyzed using four liquid chromatography coupled with high resolution mass spectrometry (LC–MS) protocols, either in reversed or normal-phase and with both types of ionization. The performances of each method were compared according to putative metabolite coverage, method repeatability and also extraction parameters such as overlap, linearity and matrix efect; in both untargeted (global) and targeted approaches using fifty standard spiked analytes.
Results: Our results verifed the broad specifcity and outstanding accuracy of solvent precipitation, namely methanol and methanol/acetonitrile. We also reveal high orthogonality between methanol-based methods and SPE, providing the possibility of increased metabolome coverage, however we highlight that such potential benefts must be weighed against time constrains, sample consumption and the risk of low reproducibility of SPE method. Furthermore, we highlighted the careful consideration about matrix choice. Plasma showed the most suitable in this metabolomics approach combined with methanol-based methods.
Conclusions: Our work proposes to facilitate rational design of protocols towards standardization of these approaches to improve the impact of metabolomics research.
Keywords Metabolomic, Guideline, Extraction, Methods, Suitability
Address and Contact Information 1 Inserm Unit IRMETIST, UMR U1313, University of Poitiers, Faculty of Medicine and Pharmacy, 86073 Poitiers, France
2 IC2MP: Institut de Chimie des Milieux et Materiaux de Poitiers, Chemistry, France
3 Cardio‐Thoracic and Vascular Surgery Intensive Care Unit, Coordination of P.M.O. CHU Poitiers, 86021 Poitiers, France
4 Biochemistry Department CHU Poitiers, 86021 Poitiers, France
5 University Hospital Federation SUPPORT Tours Poitiers Limoges, 86021 Poitiers, France
*Corresponding author: raphael.thuillier@univ-poitiers.fr
Read full article at BMC

No.  44DOI: 10.1186/s11658-023-00451-y Volume 28 (2023) - 28:44
Title AN INTEGRATIVE REVIEW OF NONOBVIOUS PUZZLES OF CELLULAR AND MOLECULAR CARDIOONCOLOGY
Authors Paweł Uruski1, Julia Matuszewska2, Aleksandra Leśniewska2, Daniel Rychlewski2, Arkadiusz Niklas1, Justyna Mikuła‐Pietrasik2, Andrzej Tykarski1 and Krzysztof Książek2*
Abstract Oncologic patients are subjected to four major treatment types: surgery, radiotherapy, chemotherapy, and immunotherapy. All nonsurgical forms of cancer management are known to potentially violate the structural and functional integrity of the cardiovascular system. The prevalence and severity of cardiotoxicity and vascular abnormalities led to the emergence of a clinical subdiscipline, called cardiooncology. This relatively new, but rapidly expanding area of knowledge, primarily focuses on clinical observations linking the adverse efects of cancer therapy with deteriorated quality of life of cancer survivors and their increased morbidity and mortality. Cellular and molecular determinants of these relations are far less understood, mainly because of several unsolved paths and contradicting findings in the literature. In this article, we provide a comprehensive view of the cellular and molecular etiology of cardiooncology. We pay particular attention to various intracellular processes that arise in cardiomyocytes, vascular endothelial cells, and smooth muscle cells treated in experimentally-controlled conditions in vitro and in vivo with ionizing radiation and drugs representing diverse modes of anti-cancer activity.
Keywords Cardiooncology, Cardiac cells, Cardiotoxicity, Vascular cells
Address and Contact Information 1 Department of Hypertensiology, Poznań University of Medical Sciences, Długa 1⁄2 Str., 61‐848 Poznan, Poland
2 Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa 1⁄2 Str., 61‐848 Poznan, Poland
*Corresponding author: kksiazek@ump.edu.pl
Read full article at BMC

No.  45DOI: 10.1186/s11658-023-00455-8 Volume 28 (2023) - 28:45
Title DELETION OF CD36 EXHIBITS LIMITED IMPACT ON NORMAL HEMATOPOIESIS AND THE LEUKEMIA MICROENVIRONMENT
Authors Yiting Meng1, Mateusz Pospiech1, Atham Ali1, Ritu Chandwani2, Mary Vergel3, Sandra Onyemaechi2, George Yaghmour4,5, Rong Lu3 and Houda Alachkar1,5*
Abstract Background: CD36 has been identifed as a potential therapeutic target both in leukemic cells and in the tumor immune microenvironment. In acute myeloid leukemia (AML), we found that APOC2 acts with CD36 to promote leukemia growth by activating the LYN-ERK signaling. CD36 also plays a role in lipid metabolism of cancer associated T-cells leading to impaired cytotoxic CD8+ T-cell and enhanced Treg cell function. To establish CD36 as a viable therapeutic target in AML, we investigated whether targeting CD36 has any detrimental impact on normal hematopoietic cells.
Methods: Diferential expression data of CD36 during human and mouse normal hematopoiesis were examined and compared. Cd36 knockout (Cd36-KO) mice were evaluated for blood analysis, hematopoietic stem cells and progenitors (HSPCs) function and phenotype analyses, and T cells in vitro expansion and phenotypes in comparison with wild type (WT) mice. In addition, MLL-PTD/FLT3-ITD leukemic cells were engrafted into Cd36-KO and WT mice, and leukemia burden was compared between groups.
Results: RNA-Seq data showed that Cd36 expression was low in HSPCs and increased as cells matured. Phenotypic analysis revealed limited changes in blood count except for a slight yet signifcantly lower red blood cell count and hemoglobin and hematocrit levels in Cd36-KO mice compared with WT mice (P<0.05). In vitro cell proliferation assays of splenocytes and HSPCs from Cd36-KO mice showed a similar pattern of expansion to that of cells from WT mice. Characterization of HSPCs showed similar percentages of the diferent progenitor cell populations between Cd36-KO with WT mice. However, Cd36-KO mice exhibited~40% reduction of the number of colonies developed from HSPCs cells compared with WT mice (P<0.001). Cd36-KO and WT mice presented comparably healthy BM transplant in non-competitive models and developed similar leukemia burden.
Conclusions: Although the loss of Cd36 afects the hematopoietic stem cell and erythropoiesis, limited detrimental overall impact was observed on normal Hematopoietic and leukemic microenvironments. Altogether, considering the limited impact on normal hematopoiesis, therapeutic approaches to target CD36 in cancer are unlikely to result in toxicity to normal blood cells.
Keywords CD36, Knockout, Hematopoiesis, T Cells, Acute myeloid leukemia, Mouse
Address and Contact Information 1 Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
2 Department of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
4 Division of Hematology, Department of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
5 USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
*Corresponding author: alachkar@usc.edu
Read full article at BMC

No.  46DOI: 10.1186/s11658-023-00458-5 Volume 28 (2023) - 28:46
Title Hsp70-Bim INTERACTION FACILITATES MITOPHAGY BY RECRUITING PARKIN AND TOMM20 INTO A COMPLEX
Authors Ting Song1*, Fangkui Yin1, Ziqian Wang1, Hong Zhang2, Peng Liu2, Yafei Guo2, Yao Tang2 and Zhichao Zhang1*
Abstract Background: For cancer therapy, the identifcation of both selective autophagy targets and small molecules that specifcally regulate autophagy is greatly needed. Heat shock protein 70 (Hsp70) is a recently discovered BH3 receptor that forms a protein‒protein interaction (PPI) with Bcl-2-interacting mediator of cell death (Bim). Herein, a specifc inhibitor of the Hsp70-Bim PPI, S1g-2, and its analog S1, which is a Bcl-2-Bim disruptor, were used as chemical tools to explore the role of Hsp70-Bim PPI in regulating mitophagy.
Methods: Co-immunoprecipitation and immunofuorescence assays were used to determine protein interactions and colocalization patterns. Organelle purifcation and immunodetection of LC3-II/LC3-I on mitochondria, endoplasmic reticulum (ER) and Golgi were applied to identify specifc types of autophagy. Cell-based and in vitro ubiquitination studies were used to study the role of the Hsp70-Bim PPI in parkin-mediated ubiquitination of outer mitochondrial membrane 20 (TOMM20).
Results: We found that after the establishment of their PPI, Hsp70 and Bim form a complex with parkin and TOMM20, which in turn facilitates parkin translocation to mitochondria, TOMM20 ubiquitination and mitophagic flux independent of Bax/Bak. Moreover, S1g-2 selectively inhibits stress-induced mitophagy without interfering with basal autophagy.
Conclusions: The findings highlight the dual protective function of the Hsp70-Bim PPI in regulating both mitophagy and apoptosis. S1g-2 is thus a newly discovered antitumor drug candidate that drives both mitophagy and cell death via apoptosis.
Keywords Apoptosis, Hsp70-Bim, Mitophagy, Parkin, TOMM20
Address and Contact Information 1 State Key Laboratory of Fine Chemicals, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning, China
2 School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
*Corresponding author: songting@dlut.edu.cn; zczhang@dlut.edu.cn
Read full article at BMC

No.  47DOI: 10.1186/s11658-023-00461-w Volume 28 (2023) - 28:47
Title AN Fc-MUTED BISPECIFIC ANTIBODY TARGETING PD-L1 AND 4-1BB INDUCES ANTITUMOR IMMUNE ACTIVITY IN COLORECTAL CANCER WITHOUT SYSTEMIC TOXICITY
Authors Lian‐sheng Cheng1,2,3,4†, Min Zhu5†, Yan Gao5†, Wen‐ting Liu2†, Wu Yin1,3, Pengfei Zhou2, Zhongliang Zhu5, Liwen Niu5, Xiaoli Zeng2, Dayan Zhang2, Qing Fang2, Fengrong Wang2, Qun Zhao2, Yan Zhang6* and Guodong Shen1,3*
Abstract Background Resistance to immune checkpoint inhibitor (ICI) therapy narrows the efficacy of cancer immunotherapy. Although 4-1BB is a promising drug target as a costimulatory molecule of immune cells, no 4-1BB agonist has been given clinical approval because of severe liver toxicity or limited efficacy. Therefore, a safe and efficient immunostimulatory molecule is urgently needed for cancer immunotherapy.
Methods HK010 was generated by antibody engineering, and the Fab/antigen complex structure was analyzed using crystallography. The affinity and activity of HK010 were detected by multiple in vitro bioassays, including enzyme-linked immunosorbent assay (ELISA), surface plasmon resonance (SPR), flow cytometry, and luciferase-reporter assays. Humanized mice bearing human PD-L1-expressing MC38 (MC38/hPDL1) or CT26 (CT26/hPDL1) tumor transplants were established to assess the in vivo antitumor activity of HK010. The pharmacokinetics (PK) and toxicity of HK010 were evaluated in cynomolgus monkeys.
Results HK010 was generated as an Fc-muted immunoglobulin (Ig)G4 PD-L1x4-1BB bispecific antibody (BsAb) with a distinguished Fab/antigen complex structure, and maintained a high affinity for human PD-L1 (KD: 2.27 nM) and low affinity for human 4-1BB (KD: 493 nM) to achieve potent PD-1/PD-L1 blockade and appropriate 4-1BB agonism. HK010 exhibited synergistic antitumor activity by blocking the PD-1/PD-L1 signaling pathway and stimulating the 4-1BB signaling pathway simultaneously, and being strictly dependent on the PD-L1 receptor in vitro and in vivo. In particular, when the dose was decreased to 0.3 mg/kg, HK010 still showed a strong antitumor effect in a humanized mouse model bearing MC38/hPDL1 tumors. Strikingly, HK010 treatment enhanced antitumor immunity and induced durable antigen-specific immune memory to prevent rechallenged tumor growth by recruiting CD8+ T cells and other lymphocytes into tumor tissue and activating tumor-infiltrating lymphocytes. Moreover, HK010 not only did not induce nonspecific production of proinflammatory cytokines but was also observed to be well tolerated in cynomolgus monkeys in 5 week repeated-dose (5, 15, or 50 mg/kg) and single-dose (75 or 150 mg/kg) toxicity studies.
Conclusion We generated an Fc-muted anti-PD-L1x4-1BB BsAb, HK010, with a distinguished structural interaction with PD-L1 and 4-1BB that exhibits a synergistic antitumor effect by blocking the PD-1/PD-L1 signaling pathway and stimulating the 4-1BB signaling pathway simultaneously. It is strictly dependent on the PD-L1 receptor with no systemic toxicity, which may offer a new option for cancer immunotherapy.
Keywords Colorectal cancer, Cancer immunotherapy, 4-1BB/CD137, PD-1/PD-L1, Immune checkpoint inhibitor, Antitumor immunity
Address and Contact Information 1 Department of Geriatrics, The First Afliated Hospital of University of Science and Technology of China, Gerontology Institute of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui, China.
2 Hefei HankeMab Biotechnology Limited, Hefei 230088, Anhui, China.
3 Anhui Provincial Key Laboratory of Tumor Immunotherapy and Nutrition Therapy, Hefei 230001, Anhui, China.
4 Anhui Province Key Laboratory of Gene Engineering Pharmaceutical, Biomedicine Technology Innovation Center of Hefei, Anhui Anke Biotechnology (Group) Co., Ltd., Hefei 230088, Anhui, China.
5 School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China.
6 School of Health Service Management, Anhui Medical University, Hefei 230032, Anhui, China.
*Corresponding author: zhangymail@ahmu.edu.cn; gdshen@ustc.edu.cn
Lian-sheng Cheng, Min Zhu, Yan Gao, and Wen-ting Liu have contributed equally to this work.
Read full article at BMC

No.  48DOI: 10.1186/s11658-023-00460-x Volume 28 (2023) - 28:48
Title PRDX1 NEGATIVELY REGULATES BLEOMYCIN-INDUCED PULMONARY FBROSIS VIA INHIBITING THE EPITHELIAL-MESENCHYMAL TRANSITION AND LUNG FBROBLAST PROLIFERATION IN VITRO AND IN VIVO
Authors Hu‐Nan Sun1*†, Chen‐Xi Ren1†, Dong Hun Lee2†, Wei‐Hao Wang3, Xiao‐Yu Guo1, Ying‐Ying Hao1, Xiao‐Ming Wang4, Hui‐Na Zhang1, Wan‐Qiu Xiao1, Nan Li1, Jie Cong1, Ying‐Hao Han1* and Taeho Kwon5,6*
Abstract Background Pulmonary fibrosis is a major category of end-stage changes in lung diseases, characterized by lung epithelial cell damage, proliferation of fibroblasts, and accumulation of extracellular matrix. Peroxiredoxin 1 (PRDX1), a member of the peroxiredoxin protein family, participates in the regulation of the levels of reactive oxygen species in cells and various other physiological activities, as well as the occurrence and development of diseases by functioning as a chaperonin.
Methods Experimental methods including MTT assay, morphological observation of fibrosis, wound healing assay, fluorescence microscopy, flow cytometry, ELISA, western blot, transcriptome sequencing, and histopathological analysis were used in this study.
Results PRDX1 knockdown increased ROS levels in lung epithelial cells and promoted epithelial-mesenchymal transition (EMT) through the PI3K/Akt and JNK/Smad signalling pathways. PRDX1 knockout significantly increased TGF-β secretion, ROS production, and cell migration in primary lung fibroblasts. PRDX1 deficiency also increased cell proliferation, cell cycle circulation, and fibrosis progression through the PI3K/Akt and JNK/Smad signalling pathways. BLM treatment induced more severe pulmonary fibrosis in PRDX1-knockout mice, mainly through the PI3K/Akt and JNK/Smad signalling pathways.
Conclusions Our findings strongly suggest that PRDX1 is a key molecule in BLM-induced lung fibrosis progression and acts through modulating EMT and lung fibroblast proliferation; therefore, it may be a therapeutic target for the treatment of BLM-induced lung fibrosis.
Keywords Pulmonary fibrosis, Peroxiredoxin 1, Reactive oxygen species, Epithelial- mesenchymal transition, Cell proliferation, PI3K/Akt and JNK/Smad signalling pathways
Address and Contact Information 1 Stem Cell and Regenerative Biology Laboratory, College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Xingyang Road #2, Daqing 163319, Heilongjiang, China.
2 Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, 77 Yongbong‐Ro, Buk‐Gu, Gwangju 61186, Republic of Korea.
3 National Coarse Cereals Engineering Research Center, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
4 Yabian Academy of Agricultural Science, Longjing, Jilin 1334000, China.
5 Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 351‐33 Neongme‐Gil, Ibam‐Myeon, Jeongeup‐Si, Jeonbuk 56216, Republic of Korea.
6 Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Dae‐jeon 34113, Republic of Korea.
*Corresponding author: sunhunan76@163.com; hyhbynd@163.com; kwon@kribb.re.kr
Hu-Nan Sun, Chen-Xi Ren and Dong Hun Lee contributed equally to this work.
Read full article at BMC

No.  49DOI: 10.1186/s11658-023-00463-8 Volume 28 (2023) - 28:49
Title A NOVEL TYROSINE tRNA-DERIVED FRAGMENT, tRFTyr, INDUCES ONCOGENESIS AND LACTATE ACCUMULATION IN LSCC BY INTERACTING WITH LDHA
Authors Rui Zhao1, Zhenming Yang1, Bo Zhao2, Wenjing Li1, Yaohui Liu1, Xiaoxue Chen1, Jing Cao1, Jiarui Zhang1, Yan Guo1, Licheng Xu1, Jinpeng Wang1, Yanan Sun1, Ming Liu1* and Linli Tian1*
Abstract Background Transfer (t)RNA-derived small RNA (tsRNA), generated from precursor or mature tRNA, is a new type of small non-coding RNA (sncRNA) that has recently been shown to play a vital role in human cancers. However, its role in laryngeal squamous cell carcinoma (LSCC) remains unclear.
Methods We elucidated the expression profiles of tsRNAs in four paired LSCC and non-neoplastic tissues by sequencing and verified the sequencing data by quantitative real-time PCR (qRT–PCR) of 60 paired samples. The tyrosine-tRNA derivative tRFTyr was identified as a novel oncogene in LSCC for further study. Loss-of-function experiments were performed to evaluate the roles of tRFTyr in tumorigenesis of LSCC. Mechanistic experiments including RNA pull-down, parallel reaction monitoring (PRM) and RNA immunoprecipitation (RIP) were employed to uncover the regulatory mechanism of tRFTyr in LSCC.
Results tRFTyr was significantly upregulated in LSCC samples. Functional assays showed that knockdown of tRFTyr significantly suppressed the progression of LSCC. A series of mechanistic studies revealed that tRFTyr could enhance the phosphorylated level of lactate dehydrogenase A (LDHA) by interacting with it. The activity of LDHA was also activated, which induced lactate accumulation in LSCC cells.
Conclusions Our data delineated the landscape of tsRNAs in LSCC and identified the oncogenic role of tRFTyr in LSCC. tRFTyr could promote lactate accumulation and tumour progression in LSCC by binding to LDHA. These findings may aid in the development of new diagnostic biomarkers and provide new insights into therapeutic strategies for LSCC.
Keywords tsRNAs, tRFTyr, LDHA, Lactate, LSCC
Address and Contact Information 1 Department of Otolaryngology-Head and Neck Surgery, The Second Afliated Hospital of Harbin Medical University, Harbin, China
2 School of Forestry, Northeast Forestry University, Harbin, China
*Corresponding author: liumingorl@qq.com; tianlinli78@163.com
Read full article at BMC

No.  50DOI: 10.1186/s11658-023-00459-4 Volume 28 (2023) - 28:50
Title NOTCH MEDIATES THE GLYCOLYTIC SWITCH VIA PI3K/Akt SIGNALING TO SUPPORT EMBRYONIC DEVELOPMENT
Authors Heng Wang1†, Wenqi Liang1†, Xuyang Wang1, Yuchun Zhan1, Wence Wang1, Lin Yang1* and Yongwen Zhu1*
Abstract Background Energy metabolism disorder or insufficient energy supply during incubation will affect the development and survival of avian embryos. Especially, β-oxidation could not provide the continuous necessary energy for avian embryonic development due to the increasing energy demand under hypoxic conditions during the mid–late embryonic stages. The role and mechanism of hypoxic glycolysis replacing β-oxidation as the main source of energy supply for avian embryonic development in the mid–late stages is unclear.
Results Here, we found that in ovo injection with glycolysis inhibitor or γ-secretase inhibitor both decreased the hepatic glycolysis level and impaired goose embryonic development. Intriguingly, the blockade of Notch signaling is also accompanied by the inhibition of PI3K/Akt signaling in the embryonic primary hepatocytes and embryonic liver. Notably, the decreased glycolysis and impaired embryonic growth induced by the blockade of Notch signaling were restored by activation of PI3K/Akt signaling.
Conclusions Notch signaling regulates a key glycolytic switch in a PI3K/Akt-dependent manner to supply energy for avian embryonic growth. Our study is the first to demonstrate the role of Notch signaling-induced glycolytic switching in embryonic development, and presents new insight into the energy supply patterns in embryogenesis under hypoxic conditions. In addition, it may also provide a natural hypoxia model for developmental biology studies such as immunology, genetics, virology, cancer, etc.
Highlights
- Glycolysis plays a dominant role in the energy supply to support mid–late avian embryonic growth.
- Notch signaling regulates a switch to glycolysis in the liver.
- Notch switched glycolysis in a PI3K/Akt-dependent manner in vivo and in vitro.
Keywords Notch signaling, Glycolysis, PI3K/Akt, Avian embryonic development
Address and Contact Information 1 Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510000, China
*Corresponding author: 2465169337@qq.com; 408034085@qq.com
Heng Wang and Wenqi Liang contributed equally to this work.
Read full article at BMC

No.  51DOI: 10.1186/s11658-023-00462-9 Volume 28 (2023) - 28:51
Title THE NLRP3 INFLAMMASOME: CONTRIBUTIONS TO INFLAMMATION-RELATED DISEASES
Authors Ying Chen1†, Xingyan Ye2,3†, Germaine Escames4,5,6†, Wangrui Lei2,3, Xin Zhang7, Meng Li7, Tong Jing7, Yu Yao2,3, Zhenye Qiu2,3, Zheng Wang8, Darío Acuña‐Castroviejo4,5,6* and Yang Yang2,3*
Abstract The NOD-like receptor protein 3 (NLRP3) inflammasome is a protein complex that regulates innate immune responses by activating caspase-1 and the inflammatory cytokines interleukin (IL)-1β and IL-18. Multiple studies have demonstrated the importance of the NLRP3 inflammasome in the development of immune and inflammation-related diseases, including arthritis, Alzheimer’s disease, inflammatory bowel disease, and other autoimmune and autoinflammatory diseases. This review first explains the activation and regulatory mechanism of the NLRP3 inflammasome. Secondly, we focus on the role of the NLRP3 inflammasome in various inflammation-related diseases. Finally, we look forward to new methods for targeting the NLRP3 inflammasome to treat inflammation-related diseases, and provide new ideas for clinical treatment.
Keywords NLRP3 infammasome, Innate immunity, Infammatory disease
Address and Contact Information 1 Department of Hematology, The First Afliated Hospital of Xi’an Jiaotong University, Xi’an, China
2 Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi’an, China
3 Department of Neurology, Xi’an No. 3 Hospital, The Afliated Hospital of Northwest University, Xi’an, China
4 Biomedical Research Center, Health Sciences Technology Park, University of Granada, Avda. del Conocimiento s/n, Granada, Spain
5 Ibs. Granada and CIBERfes, Granada, Spain
6 UGC of Clinical Laboratories, University San Cecilio’s Hospital, Granada, Spain
7 Department of Cardiology, Afliated Hospital, Yan’an University, Yan’an, China
8 Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People’s Liberation Army, Wuhan, China
*Corresponding author: dacuna@ugr.es; yang200214yy@nwu.edu.cn
Ying Chen, Xingyan Ye, and Germaine Escames contributed equally to this work.
Read full article at BMC

No.  52DOI: 10.1186/s11658-023-00464-7 Volume 28 (2023) - 28:52
Title CKM AND TERT DUAL PROMOTERS DRIVE CRISPR–dCas9 TO SPECIFCALLY INHIBIT THE MALIGNANT BEHAVIOR OF OSTEOSARCOMA CELLS
Authors Yawei Hu1, Hao Zhang1, Zengfeng Guo1, Jianhua Zhou1, Wang Zhang1, Ming Gong1 and Jiawen Wu1*
Abstract Improvements in treatment and chemotherapy have increased the survival rate of osteosarcoma, but overall efficacy remains low, highlighting the need for new gene therapy methods. Clustered regularly interspaced short palindromic repeats–deactivated Cas9 (CRISPR–dCas9) technology offers a promising strategy, but targeting osteosarcoma cells precisely is a challenge. We designed a system to achieve specific expression of CRISPR–dCas9–KRAB in osteosarcoma cells by using the creatine kinase muscle (CKM) promoter to drive dCas9–KRAB and the telomerase reverse transcriptase (TERT) promoter to drive single guide (sg)RNA expression. We inhibited the MDM2 proto-oncogene using this system in vitro, which efficiently inhibited the malignant behavior of osteosarcoma cells and induced apoptosis without affecting normal cells. In vivo experiments demonstrated that this system effectively inhibited the growth of subcutaneously transplanted tumors in nude mice. These findings provide a new method for precise identification and intervention of osteosarcoma with significant implications for the development of gene therapy methods for other cancers. Future research should focus on optimizing this system for clinical translation.
Keywords Osteosarcoma, CKM promoter, TERT promoter, CRISPR–dCas9 system, Gene therapy
Address and Contact Information 1 Department of Spine Surgery, People’s Hospital of Longhua, Affliated Hospital of Southern Medical University, Shenzhen, China
*Corresponding author: wujiawen_80@126.com
Read full article at BMC

No.  53DOI: 10.1186/s11658-023-00471-8 Volume 28 (2023) - 28:53
Title EFEMP2 UPREGULATES PD‐L1 EXPRESSION VIA EGFR/ERK1/2/c‐Jun SIGNALING TO PROMOTE THE INVASION OF OVARIAN CANCER CELLS
Authors Xin Shen1†, Xuli Jin2†, Shuang Fang3 and Jie Chen1*
Abstract Background Fibulin-like extracellular matrix protein 2 (EFEMP2) has been reported to be related to the progression of various cancers. We have previously reported that EFEMP2 was highly expressed in ovarian cancer and was strongly associated with poor prognosis in patients. This study intends to further explore its interacting proteins and possible downstream signaling pathways.
Method The expression of EFEMP2 was detected by RT-qPCR, ICC and western blot in 4 kinds of ovarian cancer cells with different migration and invasion ability. Cell models with strong or weak EFEMP2 expression were constructed by lentivirus transfection. The effects of the down-regulation and up-regulation of EFEMP2 on the biological behavior of ovarian cancer cells were studied through in-vitro and in-vivo functional tests. The phosphorylation pathway profiling array and KEGG database analyses identified the downstream EGFR/ERK1/2/c-Jun signaling pathway and the programmed death-1 (PD-L1) pathway enrichment. Additionally, the protein interaction between EFEMP2 and EGFR was detected by
immunoprecipitation. Result EFEMP2 was positively correlated with the invasion ability of ovarian cancer cells, its down-regulation inhibited the migrative, invasive and cloning capacity of cancer cells in vitro and suppressed the tumor proliferation and intraperitoneal diffusion in vivo, while its up-regulation did the opposite. Moreover, EFEMP2 could bind to EGFR to induce PD-L1 regulation in ovarian cancer, which was caused by the activation of EGFR/ERK1/2/c-Jun signaling. Similar to EFEMP2, PD-L1 was also highly expressed in aggressive cells and had the ability to promote the invasion and metastasis of ovarian cancer cells both in vitro and in vivo, and PD-L1 upregulation was partly caused by EFEMP2 activation. Afatinib combined with trametinib had an obvious effect of inhibiting the intraperitoneal diffusion of ovarian cancer cells, especially in the group with low expression of EFEMP2, while overexpression of PD-L1 could reverse this phenomenon.
Conclusion EFEMP2 could bind to EGFR to activate ERK1/2/c-Jun pathway and regulate PD-L1 expression, furthermore PD-L1 was extremely essential for EFEMP2 to promote ovarian cancer cells invasion and dissemination in vitro and in vivo. Targeted therapy against the source gene EFEMP2 is our future research direction, which may better inhibit the invasion and metastasis of ovarian cancer cells.
Keywords Ovarian cancer, EFEMP2, PD-L1, EGFR, ERK1/2/c-Jun pathway, Trametinib, Afatinib
Address and Contact Information 1 Department of Maternal and Child Health, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
2 Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
3 Jinan Medical Center Management Committee, Jinan 250000, China *Corresponding author: 77chenjie@sdu.edu.cn
Xin Shen and Xuli Jin contributed equally to this work.
Read full article at BMC

No.  54DOI: 10.1186/s11658-023-00470-9 Volume 28 (2023) - 28:54
Title γ-2 AND GSG1L BIND WITH COMPARABLE AFNITIES TO THE TETRAMERIC GluA1 CORE
Authors Chenlu Yu1,2, Hendrik F. P. Runge2, Antara Mukhopadhyay1,2, Gerd Zolles3 and Maximilian H. Ulbrich1,2*
Abstract Background The AMPA-type ionotropic glutamate receptor mediates fast excitatory neurotransmission in the brain. A variety of auxiliary subunits regulate its gating properties, assembly, and trafficking, but it is unknown if the binding of these auxiliary subunits to the receptor core is dynamically regulated. Here we investigate the interplay of the two auxiliary subunits γ-2 and GSG1L when binding to the AMPA receptor composed of four GluA1 subunits.
Methods We use a three-color single-molecule imaging approach in living cells, which allows the direct observation of the receptors and both auxiliary subunits. Colocalization of different colors can be interpreted as interaction of the respective receptor subunits.
Results Depending on the relative expression levels of γ-2 and GSG1L, the occupancy of binding sites shifts from one auxiliary subunit to the other, supporting the idea that they compete for binding to the receptor. Based on a model where each of the four binding sites at the receptor core can be either occupied by γ-2 or GSG1L, our experiments yield apparent dissociation constants for γ-2 and GSG1L in the range of 2.0–2.5/µm2.
Conclusions The result that both binding affinities are in the same range is a prerequisite for dynamic changes of receptor composition under native conditions.
Keywords Receptor assembly, Subunit stoichiometry, AMPA receptor regulatory subunits, Single-molecule imaging
Address and Contact Information 1 Internal Medicine IV, Department of Medicine, University Medical Center, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
2 BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
3 Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
*Corresponding author: max.ulbrich@bioss.uni-freiburg.de
Read full article at BMC

No.  55DOI: 10.1186/s11658-023-00466-5 Volume 28 (2023) - 28:55
Title HYPOTHERMIC OXYGENATED PERFUSION ATTENUATES DCD LIVER ISCHEMIA–REPERFUSION INJURY BY ACTIVATING THE JAK2/STAT3/HAX1 PATHWAY TO REGULATE ENDOPLASMIC RETICULUM STRESS
Authors Pengpeng Yue1, Xiaoyan Lv2, Jian You1, Yongkang Zou1, Jun luo1, Zhongshan Lu1, Hankun Cao1, Zhongzhong Liu1, Xiaoli Fan1* and Qifa Ye1,3*
Abstract Background Hepatic ischemia–reperfusion injury (IRI) in donation after cardiac death (DCD) donors is a major determinant of transplantation success. Endoplasmic reticulum (ER) stress plays a key role in hepatic IRI, with potential involvement of the Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) pathway and the antiapoptotic protein hematopoietic-lineage substrate-1-associated protein X-1 (HAX1). In this study, we aimed to investigate the effects of hypothermic oxygenated perfusion (HOPE), an organ preservation modality, on ER stress and apoptosis during hepatic IRI in a DCD rat model.
Methods To investigate whether HOPE could improve IRI in DCD livers, levels of different related proteins were examined by western blotting and quantitative real-time polymerase chain reaction. Further expression analyses, immunohistochemical analyses, immunofluorescence staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining, and transmission electron microscopy were conducted to analyze the effects of HOPE on ER stress and apoptosis. To clarify the role of the JAK2/STAT3 pathway and HAX1 in this process, AG490 inhibitor, JAX1 plasmid transfection, co-immunoprecipitation (CO-IP), and flow cytometry analyses were conducted.
Results HOPE reduced liver injury and inflammation while alleviating ER stress and apoptosis in the DCD rat model. Mechanistically, HOPE inhibited unfolded protein responses by activating the JAK2/STAT3 pathway, thus reducing ER stress and apoptosis. Moreover, the activated JAK2/STAT3 pathway upregulated HAX1, promoting the interaction between HAX1 and SERCA2b to maintain ER calcium homeostasis. Upregulated HAX1 also modulated ER stress and apoptosis by inhibiting the inositol-requiring enzyme 1 (IRE1) pathway.
Conclusions JAK2/STAT3-mediated upregulation of HAX1 during HOPE alleviates hepatic ER stress and apoptosis, indicating the JAK2/STAT3/HAX1 pathway as a potential target for IRI management during DCD liver transplantation.
Keywords Liver transplantation, DCD, HOPE, IRI, JAK2/STAT3, HAX1, Endoplasmic reticulum stress
Address and Contact Information 1 Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-based Medical Materials, 430071 Wuhan, China
2 Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
3 The Third Xiangya Hospital of Central South University, Research Center of National Health Ministry On Transplantation Medicine Engineering and Technology, Changsha 410013, China
*Corresponding author: fanxl_whu@hotmail.com; yqf_china@163.com
Read full article at BMC

No.  56DOI: 10.1186/s11658-023-00473-6 Volume 28 (2023) - 28:56
Title TALIN2 AND KANK2 FUNCTIONALLY INTERACT TO REGULATE MICROTUBULE DYNAMICS, PACLITAXEL SENSITIVITY AND CELL MIGRATION IN THE MDA-MB-435S MELANOMA CELL LINE
Authors Marija Lončarić1, Nikolina Stojanović1, Anja Rac‐Justament1, Kaatje Coopmans1, Dragomira Majhen1, Jonathan D. Humphries2, Martin J. Humphries3 and Andreja Ambriović‐Ristov1*
Abstract Background Focal adhesions (FAs) are integrin-containing, multi-protein structures that link intracellular actin to the extracellular matrix and trigger multiple signaling pathways that control cell proliferation, differentiation, survival and motility. Microtubules (MTs) are stabilized in the vicinity of FAs through interaction with the components of the cortical microtubule stabilizing complex (CMSC). KANK (KN motif and ankyrin repeat domains) family proteins within the CMSC, KANK1 or KANK2, bind talin within FAs and thus mediate actin-MT crosstalk. We previously identified in MDA-MB-435S cells, which preferentially use integrin αVβ5 for adhesion, KANK2 as a key molecule enabling the actin-MT crosstalk. KANK2 knockdown also resulted in increased sensitivity to MT poisons, paclitaxel (PTX) and vincristine and reduced migration. Here, we aimed to analyze whether KANK1 has a similar role and to distinguish which talin isoform binds KANK2.
Methods The cell model consisted of human melanoma cell line MDA-MB-435S and stably transfected clone with decreased expression of integrin αV (3αV). For transient knockdown of talin1, talin2, KANK1 or KANK2 we used gene-specific siRNAs transfection. Using previously standardized protocol we isolated integrin adhesion complexes. SDS-PAGE and Western blot was used for protein expression analysis. The immunofluorescence analysis and live cell imaging was done using confocal microscopy. Cell migration was analyzed with Transwell Cell Culture Inserts. Statistical analysis using GraphPad Software consisted of either one-way analysis of variance (ANOVA), unpaired Student’s t-test or two-way ANOVA analysis.
Results We show that KANK1 is not a part of the CMSC associated with integrin αVβ5 FAs and its knockdown did not affect the velocity of MT growth or cell sensitivity to PTX. The talin2 knockdown mimicked KANK2 knockdown i.e. led to the perturbation of actin-MT crosstalk, which is indicated by the increased velocity of MT growth and increased sensitivity to PTX and also reduced migration.
Conclusion We conclude that KANK2 functionally interacts with talin2 and that the mechanism of increased sensitivity to PTX involves changes in microtubule dynamics. These data elucidate a cell-type-specific role of talin2 and KANK2 isoforms and we propose that talin2 and KANK2 are therefore potential therapeutic targets for improved cancer therapy.
Keywords Focal adhesion, Talin1, Talin2, KANK1, KANK2, Cortical microtubule stabilizing complex
Address and Contact Information 1 Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
2 Department of Life Science, Manchester Metropolitan University, Manchester, United Kingdom
3 Wellcome Centre for Cell‐Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
*Corresponding author: Andreja.Ambriovic.Ristov@irb.hr
Read full article at BMC

No.  57DOI: 10.1186/s11658-023-00453-w Volume 28 (2023) - 28:57
Title MECHANICAL FORCE INDUCES MITOPHAGY-MEDIATED ANAEROBIC OXIDATION IN PERIODONTAL LIGAMENT STEM CELLS
Authors Zijie Zhang1, Shuyue Cui1, Yajing Fu1, Jixiao Wang1, Jiani Liu1 and Fulan Wei1*
Abstract Background The preference for glucose oxidative mode has crucial impacts on various physiological activities, including determining stem cell fate. External mechanical factors can play a decisive role in regulating critical metabolic enzymes and pathways of stem cells. Periodontal ligament stem cells (PDLSCs) are momentous effector cells that transform mechanical force into biological signals during the reconstruction of alveolar bone. However, mechanical stimuli-induced alteration of oxidative characteristics in PDLSCs and the underlying mechanisms have not been fully elucidated.
Methods Herein, we examined the expression of LDH and COX4 by qRT-PCR, western blot, immunohistochemistry and immunofluorescence. We detected metabolites of lactic acid and reactive oxygen species for functional tests. We used tetramethylrhodamine methyl ester (TMRM) staining and a transmission electron microscope to clarify the mitochondrial status. After using western blot and immunofluorescence to clarify the change of DRP1, we further examined MFF, PINK1, and PARKIN by western blot. We used cyclosporin A (CsA) to confirm the regulation of mitophagy and ceased the stretching as a rescue experiment.
Results Herein, we ascertained that mechanical force could increase the level of LDH and decrease the expression of COX4 in PDLSCs. Simultaneously, the yield of reactive oxygen species (ROS) in PDLSC reduced after stretching, while lactate acid augmented significantly. Furthermore, mitochondrial function in PDLSCs was negatively affected by impaired mitochondrial membrane potential (MMP) under mechanical force, and the augment of mitochondrial fission further induced PRKN-dependent mitophagy, which was confirmed by the rescue experiments via blocking mitophagy. As a reversible physiological stimulation, the anaerobic preference of PDLSCs altered by mechanical force could restore after the cessation of force stimulation.
Conclusions Altogether, our study demonstrates that PDLSCs under mechanical force preferred anaerobic oxidation induced by the affected mitochondrial dynamics, especially mitophagy. Our findings support an association between mechanical stimulation and the oxidative profile of stem cells, which may shed light on the mechanical guidance of stem cell maintenance and commitment, and lay a molecular foundation for periodontal tissue regeneration.
Keywords Mechanical force, Stem cells, Metabolism, Mitophagy
Address and Contact Information 1 Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University Cheeloo College of Medicine, No.44-1 Wenhua Road West, Jinan 250012, Shandong, China
*Corresponding author: weif@email.sdu.edu.cn
Read full article at BMC

No.  58DOI: 10.1186/s11658-023-00476-3 Volume 28 (2023) - 28:58
Title Melanoma cells induce dedifferentiation and metabolic changes in adipocytes present in the tumor niche
Authors Aleksandra Simiczyjew1*, Justyna Wądzyńska1, Katarzyna Pietraszek‐Gremplewicz1, Magdalena Kot1, Marcin Ziętek2,3, Rafał Matkowski2,3 and Dorota Nowak1
Abstract Background One of the factors that affect the progression of melanoma is the tumor microenvironment, which consists of cellular elements, extracellular matrix, acidification, and a hypoxic state. Adipocytes are one of the types of cell present in the niche and are localized in the deepest layer of the skin. However, the relationship between fat cells and melanoma remains unclear.
Methods We assessed the influence of melanoma cells on adipocytes using an indirect coculture system. We estimated the level of cancer-associated adipocyte (CAA) markers through quantitative PCR analysis. The fibroblastic phenotype of CAAs was confirmed by cell staining and western blotting analysis. The lipid content was estimated by lipid detection in CAAs using LipidSpot and by quantitative analysis using Oil Red O. The expression of proteins involved in lipid synthesis, delipidation, and metabolic processes were assessed through quantitative PCR or western blotting analysis. Lactate secretion was established using a Lactate-Glo™ assay. Proteins secreted by CAAs were identified in cytokine and angiogenesis arrays. The proliferation of melanoma cells cocultured with CAAs was assessed using an XTT proliferation assay. Statistical analysis was performed using a one-way ANOVA followed by Tukey’s test in GraphPad Prism 7 software.
Results Obtained CAAs were identified by decreased levels of leptin, adiponectin, resistin, and FABP4. Adipocytes cocultured with melanoma presented fibroblastic features, such as a similar proteolytic pattern to that of 3T3L1 fibroblasts and increased levels of vimentin and TGFβRIII. Melanoma cells led to a reduction of lipid content in CAAs, possibly by downregulation of lipid synthesis pathways (lower FADS, SC4MOL, FASN) or enhancement of lipolysis (higher level of phosphorylation of ERK and STAT3). Adipocytes cocultured with melanoma cells secreted higher IL6 and SerpinE1 levels and produced less CCL2, CXCL1, and angiogenic molecules. CAAs also showed metabolic changes comprising the increased secretion of lactate and enhanced production of glucose, lactate, and ion transporters. In addition, changes in adipocytes observed following melanoma coculture resulted in a higher proliferation rate of cancer cells.
Conclusions Melanoma cells led to decreased lipid content in adipocytes, which might be related to enhanced delipidation or reduction of lipid synthesis. Fibroblast-like CAAs showed metabolic changes that may be the reason for accelerated proliferation of melanoma cells.
Keywords Adipocytes, Melanoma, Cancer-associated adipocytes
Address and Contact Information 1 Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50‐383 Wroclaw, Poland
2 Department of Oncology and Division of Surgical Oncology, Wroclaw Medical University, Plac Hirszfelda 12, 53‐413 Wroclaw, Poland
3 Lower Silesian Oncology, Pulmonology, and Hematology Center, Plac Hirszfelda 12, 53‐413 Wroclaw, Poland
*Corresponding author: aleksandra.simiczyjew@uwr.edu.pl
Read full article at BMC

No.  59DOI: 10.1186/s11658-023-00472-7 Volume 28 (2023) - 28:59
Title GENETICALLY ENCODED PHOSPHATIDYLSERINE BIOSENSOR FOR IN VITRO, EX VIVO AND IN VIVO LABELLING
Authors Eimina Dirvelyte1, Daina Bujanauskiene1,2, Evelina Jankaityte1,3, Neringa Daugelaviciene1, Ugne Kisieliute2, Igor Nagula1, Rima Budvytyte1,3 and Urte Neniskyte1,2*
Abstract Background The dynamics of phosphatidylserine in the plasma membrane is a tightly regulated feature of eukaryotic cells. Phosphatidylserine (PS) is found preferentially in the inner leaflet of the plasma membrane. Disruption of this asymmetry leads to the exposure of phosphatidylserine on the cell surface and is associated with cell death, synaptic pruning, blood clotting and other cellular processes. Due to the role of phosphatidylserine in widespread cellular functions, an efficient phosphatidylserine probe is needed to study them. Currently, a few different phosphatidylserine labelling tools are available; however, these labels have unfavourable signal-to-noise ratios and are difficult to use in tissues due to limited permeability. Their application in living tissue requires injection procedures that damage the tissue and release damage-associated molecular patterns, which in turn stimulates phosphatidylserine exposure.
Methods For this reason, we developed a novel genetically encoded phosphatidylserine probe based on the C2 domain of the lactadherin (MFG-E8) protein, suitable for labelling exposed phosphatidylserine in various research models. We tested the C2 probe specificity to phosphatidylserine on hybrid bilayer lipid membranes by observing surface plasmon resonance angle shift. Then, we analysed purified fused C2 proteins on different cell culture lines or engineered AAVs encoding C2 probes on tissue cultures after apoptosis induction. For in vivo experiments, neurotropic AAVs were intravenously injected into perinatal mice, and after 2 weeks, brain slices were collected to observe C2-SNAP expression.
Results The biophysical analysis revealed the high specificity of the C2 probe for phosphatidylserine. The fused recombinant C2 proteins were suitable for labelling phosphatidylserine on the surface of apoptotic cells in various cell lines. We engineered AAVs and validated them in organotypic brain tissue cultures for non-invasive delivery of the genetically encoded C2 probe and showed that these probes were expressed in the brain in vivo after intravenous AAV delivery to mice.
Conclusions We have demonstrated that the developed genetically encoded PS biosensor can be utilised in a variety of assays as a two-component system of C2 and C2m2 fusion proteins. This system allows for precise quantification and PS visualisation at directly specified threshold levels, enabling the evaluation of PS exposure in both physiological and cell death processes.
Keywords Phosphatidylserine, Genetically encoded probe, Lactadherin, MFG-E8, C2 domain, SNAP-tag
Address and Contact Information 1 VU LSC-EMBL Partnership Institute for Genome Editing Technologies, Life Sciences Center, Vilnius University, Vilnius, Lithuania
2 Institute of Bioscience, Life Sciences Center, Vilnius University, Vilnius, Lithuania
3 Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
*Corresponding author: urte.neniskyte@gmc.vu.lt
Read full article at BMC

No.  60DOI: 10.1186/s11658-023-00467-4 Volume 28 (2023) - 28:60
Title THE ROLE OF NPY2R/NFATc1/DYRK1A REGULATORY AXIS IN SEBACEOUS GLANDS FOR SEBUM SYNTHESIS
Authors Tao Yang1†, Renyi Hei2†, Xiaosong Li3†, Tianhua Ma4, Yifen Shen5, Chao Liu5, Wen He5, Lin Zhu5, Yongchun Gu5, Yanping Hu6, Wenbin Wei7* and Yihang Shen5*
Abstract Background Sebaceous glands (SGs) synthesize and secret sebum to protect and moisturize the dermal system via the complicated endocrine modulation. Dysfunction of SG are usually implicated in a number of dermal and inflammatory diseases. However, the molecular mechanism behind the differentiation, development and proliferation of SGs is far away to fully understand.
Methods Herein, the rat volar and mammary tissues with abundant SGs from female SD rats with (post-natal day (PND)-35) and without puberty onset (PND-25) were arrested, and conducted RNA sequencing. The protein complex of Neuropeptide Y receptor Y2 (NPY2R)/NPY5R/Nuclear factor of activated T cells 1 (NFATc1) was performed by immunoprecipitation, mass spectrum and gel filtration. Genome-wide occupancy of NFATc1 was measured by chromatin immunoprecipitation sequencing. Target proteins’ expression and localization was detected by western blot and immunofluorescence.
Results NPY2R gene was significantly up-regulated in volar and mammary SGs of PND-25. A special protein complex of NPY2R/NPY5R/NFATc1 in PND-25. NFATc1 was dephosphorylated and activated, then localized into nucleus to exert as a transcription factor in volar SGs of PND-35. NFATc1 was especially binding at enhancer regions to facilitate the distal SG and sebum related genes’ transcription. Dual specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) contributed to NFATc1 phosphorylation in PND-25, and inactivated of DYRK1A resulted in NFATc1 dephosphorylation and nuclear localization in PND-35.
Conclusions Our findings unmask the new role of NPY2R/NFATc1/DYRK1A in pubertal SG, and are of benefit to advanced understanding the molecular mechanism of SGs’ function after puberty, and provide some theoretical basis for the treatment of acne vulgaris from the perspective of hormone regulation.
Keywords Sebaceous glands, NPY2R, NPY5R, NFATc1, DYRK1A, Puberty onset
Address and Contact Information 1 Department of Medical Cosmetology, Suzhou Ninth People’s Hospital, Suzhou 215200, Jiangsu, China.
2 Department of Otolaryngology-Head and Neck Surgery, General Hospital of Shenyang Military Area Command, Shenyang 110016, Liaoning, China.
3 Department of Anorectal Surgery, Suzhou Ninth People’s Hospital, Suzhou 215200, Jiangsu, China.
4 Graduate School of Soochow University, Suzhou 215031, Jiangsu, China.
5 Central Laboratory, Suzhou Ninth People’s Hospital, 2666, Ludang Road, Suzhou 215200, Jiangsu, China.
6 Department of Molecular Pathology, The Affliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, Henan, China.
7 Department of Oral Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology, No. 639 Zhizaoju Road, Huangpu District, Shanghai 200013, China.
*Corresponding author: tian_qian_cool@126.com; devbrother@sjtu.edu.cn
Tao Yang, Renyi Hei and Xiaosong Li contributed equally to this work.
Read full article at BMC

No.  61DOI: 10.1186/s11658-023-00468-3 Volume 28 (2023) - 28:61
Title A FROG PEPTIDE PROVIDES NEW STRATEGIES FOR THE INTERVENTION AGAINST SKIN WOUND HEALING
Authors Chao Li3†, Zhe Fu1†, Tao Jin5†, Yixiang Liu2, Naixin Liu1, Saige Yin1, Zhuo Wang3, Yubing Huang3, Yinglei Wang1, Yingxuan Zhang1, Jiayi Li1, Yutong Wu1, Li He4*, Jing Tang3*, Ying Wang2* and Xinwang Yang1*
Abstract Background Amphibian derived pro-healing peptides as molecular probes might provide a promising strategy for development of drug candidates and elucidation of cellular and molecular mechanisms of skin wound healing. A novel skin amphibian peptide, OA-RD17, was tested for modulation of cellular and molecular mechanisms associated with skin wound healing.
Methods Cell scratch, cell proliferation, trans-well, and colony formation assays were used to explore the pro-healing ability of peptide OA-RD17 and microRNA-632 (miR-632). Then, the therapeutic effects of OA-RD17 and miR-632 were assessed in mice, diabetic patient ex vivo skin wounds and SD rats. Moreover, hematoxylin and eosin (H&E), enzyme-linked immunosorbent assay (ELISA), immunohistochemistry, and immunofluorescence staining were performed to detect skin wound tissue regeneration, inflammatory factors expression, and macrophage polarization. Finally, RNA sequencing, molecular docking, co-localization, dual luciferase reporter, real-time quantitative reverse transcription PCR (RT-qPCR), and Western blotting were used to explore the mechanism of OA-RD17 and miR-632 on facilitating skin wound healing.
Results The non-toxic peptide (OA-RD17) promoted macrophage proliferation and migration by activating MAPK and suppressed inflammation by inhibiting NF-κB. In keratinocytes, OA-RD17 inhibited excessive inflammation, and activated MAPK via the Toll-like receptor 4 (TLR4) to promote proliferation and migration, as well as up-regulate the expression of miR-632, which targeted GSK3β to activate Wnt/β-catenin to boost proliferation and migration in a positive feedback manner. Notably, OA-RD17 promoted transition from the inflammatory to proliferative stage, accelerated epidermal and granulation regeneration, and exhibited therapeutic effects on mouse and diabetic patient ex vivo skin wounds. MiR-632 activated Wnt/β-catenin to promote full-thickness skin wound healing in rats.
Conclusions OA-RD17 exhibited promising therapeutic effects on mice (full-thickness, deep second-degree burns), and ex vivo skin wounds in diabetic patients by regulating macrophages proliferation, migration, and polarization (MAPK, NF-κB), and keratinocytes proliferation and migration (TLR4/MAPK/miR-632/Wnt/β-catenin molecular axis). Moreover, miR-632 also activated Wnt/β-catenin to promote full-thickness skin wound healing in rats. Notably, our results indicate that OA-RD17 and miR-632 are promising pro-healing drug candidates.
Keywords Pro-healing peptide, Amphibians, Skin wounds, TLR4/MAPK, Infammation, miR-632/Wnt/β-catenin
Address and Contact Information 1 Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
2 Key Laboratory of Chemistry in Ethnic Medicinal Resources & Key Laboratory of Natural Products Synthetic Biology of Ethnic Medicinal Endophytes, State Ethnic Afairs Commission & Ministry of Education, School of Ethnic Medicine, Yunnan Minzu University, Kunming 650504, Yunnan, China
3 Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
4 Department of Dermatology, First Afliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
5 Department of Orthopedics, 920th Hospital of Joint Logistics Support Force of PLA, Kunming 650032, Yunnan, China
*Corresponding author: drheli2662@126.com; gracett916@163.com; wangying_814@163.com; yangxinwanghp@163.com; yangxinwang@kmmu.edu.cn
Chao Li, Zhe Fu and Tao Jin are contributed equally to this manuscript.
Read full article at BMC

No.  62DOI: 10.1186/s11658-023-00465-6 Volume 28 (2023) - 28:62
Title PEPTIDASE INHIBITOR (PI16) IMPAIRS BLADDER CANCER METASTASIS BY INHIBITING NF-κB ACTIVATION VIA DISRUPTING MULTIPLE-SITE UBIQUITINATION OF NEMO
Authors Xiangqin Kuang1,2,3†, Zhuojun Zhang1,2†, Difeng Li2,9†, Wenhao Bao1,2, Jinyuan Pan4, Ping Zhou1,2, Han Chen1,2, Zhiqing Gao1,2, Xiaoyi Xie1,2, Chunxiao Yang1,2, Ge Zhu1,2, Zhongqiu Zhou5, Ruiming Tang6, Zhengfu Feng6, Lihuan Zhou6, Xiaoli Feng6, Lan Wang7, Jianan Yang1,8* and Lili Jiang1,2*
Abstract Background Bladder cancer (BLCA) is a malignancy that frequently metastasizes and leads to poor patient prognosis. It is essential to understand the molecular mechanisms underlying the progression and metastasis of BLCA and identify potential biomarkers.
Methods The expression of peptidase inhibitor 16 (PI16) was analysed using quantitative PCR, immunoblotting and immunohistochemistry assays. The functional roles of PI16 were evaluated using wound healing, transwell, and human umbilical vein endothelial cell tube formation assays, as well as in vivo tumour models. The effects of PI16 on nuclear factor κB (NF-κB) signalling activation were examined using luciferase reporter gene systems, immunoblotting and immunofluorescence assays. Co-immunoprecipitation was used to investigate the interaction of PI16 with annexin-A1 (ANXA1) and NEMO.
Results PI16 expression was downregulated in bladder cancer tissues, and lower PI16 levels correlated with disease progression and poor survival in patients with BLCA. Overexpressing PI16 inhibited BLCA cell growth, motility, invasion and angiogenesis in vitro and in vivo, while silencing PI16 had the opposite effects. Mechanistically, PI16 inhibited the activation of the NF-κB pathway by interacting with ANXA1, which inhibited K63 and M1 ubiquitination of NEMO.
Conclusions These results indicate that PI16 functions as a tumour suppressor in BLCA by inhibiting tumour growth and metastasis. Additionally, PI16 may serve as a potential biomarker for metastatic BLCA.
Keywords Bladder cancer, PI16, NF-κB, Ubiquitination
Address and Contact Information 1 Afliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China.
2 Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modifcation and Degradation, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China.
3 Department of Medical Imaging, Health Science Center, Hubei Minzu University, Enshi 445000, China.
4 Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang 438000, China.
5 Meishan Women and Children’s Hospital, Alliance Hospital of West China Second University Hospital, Sichuan University, Meishan 620000, China.
6 The Sixth Afliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou 511518, China.
7 Department of Pathogen Biology and Immunology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou 510006, China.
8 Department of Urologic Oncosurgery, Afliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China.
9 Department of Pathology, School of Medicine, Women’s Hospital, Zhejiang University, 310006 Hangzhou, China.
*Corresponding author: yangjianan@gzhmu.edu.cn; jianglili@gzhmu.edu.cn
Xiangqin Kuang, Zhuojun Zhang and Difeng Li contributed equally to this work.
Read full article at BMC

No.  6DOI: 10.1186/s11658-023-00479-0 Volume 28 (2023) - 28:63
Title MULTI-OMICS STRATEGIES UNCOVER THE MOLECULAR MECHANISMS OF NITROGEN, PHOSPHORUS AND POTASSIUM DEFICIENCY RESPONSES IN Brassica napus
Authors Ying Fu1, Annaliese S. Mason3, Maolin Song1,2, Xiyuan Ni1, Lei Liu1, Jianghua Shi1, Tanliu Wang1, Meili Xiao4, Yaofeng Zhang1, Donghui Fu4* and Huasheng Yu1*
Abstract Background Nitrogen (N), phosphorus (P) and potassium (K) are critical macronutrients in crops, such that deficiency in any of N, P or K has substantial effects on crop growth. However, the specific commonalities of plant responses to different macronutrient deficiencies remain largely unknown.
Methods Here, we assessed the phenotypic and physiological performances along with whole transcriptome and metabolomic profiles of rapeseed seedlings exposed to N, P and K deficiency stresses.
Results Quantities of reactive oxygen species were significantly increased by all macronutrient deficiencies. N and K deficiencies resulted in more severe root development responses than P deficiency, as well as greater chlorophyll content reduction in leaves (associated with disrupted chloroplast structure). Transcriptome and metabolome analyses validated the macronutrient-specific responses, with more pronounced effects of N and P deficiencies on mRNAs, microRNAs (miRNAs), circular RNAs (circRNAs) and metabolites relative to K deficiency. Tissue-specific responses also occurred, with greater effects of macronutrient deficiencies on roots compared with shoots. We further uncovered a set of common responders with simultaneous roles in all three macronutrient deficiencies, including 112 mRNAs and 10 miRNAs involved in hormonal signaling, ion transport and oxidative stress in the root, and 33 mRNAs and 6 miRNAs with roles in abiotic stress response and photosynthesis in the shoot. 27 and seven common miRNA-mRNA pairs with role in miRNA-mediated regulation of oxidoreduction processes and ion transmembrane transport were identified in all three macronutrient deficiencies. No circRNA was responsive to three macronutrient deficiency stresses, but two common circRNAs were identified for two macronutrient deficiencies. Combined analysis of circRNAs, miRNAs and mRNAs suggested that two circRNAs act as decoys for miR156 and participate in oxidoreduction processes and transmembrane transport in both N- and P-deprived roots. Simultaneously, dramatic alterations of metabolites also occurred. Associations of RNAs with metabolites were observed, and suggested potential positive regulatory roles for tricarboxylic acids, azoles, carbohydrates, sterols and auxins, and negative regulatory roles for aromatic and aspartate amino acids, glucosamine-containing compounds, cinnamic acid, and nicotianamine in plant adaptation to macronutrient deficiency.
Conclusions Our findings revealed strategies to rescue rapeseed from macronutrient deficiency stress, including reducing the expression of non-essential genes and activating or enhancing the expression of anti-stress genes, aided by plant hormones, ion transporters and stress responders. The common responders to different macronutrient deficiencies identified could be targeted to enhance nutrient use efficiency in rapeseed.
Keywords Nitrogen, Phosphorus, Potassium, Deprivation, RNA, Metabolite
Address and Contact Information 1 Institute of Crop and Nuclear Technology Utilization, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
2 College of Advanced Agricultural Sciences, Zhejiang A&F University, Hangzhou, China
3 Plant Breeding Department, University of Bonn, Katzenburgweg 5, 53115 Bonn, Germany
4 Key Laboratory of Crop Physiology, Ecology and Genetic Breeding, Ministry of Education, Agronomy College, Jiangxi Agricultural University, Nanchang 330045, China
*Corresponding author: fudhui@163.com; yuhuasheng-0@163.com
Read full article at BMC

No.  64DOI: 10.1186/s11658-023-00469-2 Volume 28 (2023) - 28:64
Title EXOGENOUS AND ENDOGENOUS dsRNAs PERCEIVED BY PLANT DICER-LIKE 4 PROTEIN IN THE RNAi-DEPLETED CELLULAR CONTEXT
Authors Paola Leonetti1†, Arianna Consiglio2†, Dennis Arendt3†, Ralph Peter Golbik3, Luisa Rubino1, Torsten Gursinsky3, Sven‐Erik Behrens3 and Vitantonio Pantaleo1*
Abstract Background In plants, RNase III Dicer-like proteins (DCLs) act as sensors of dsRNAs and process them into short 21- to 24-nucleotide (nt) (s)RNAs. Plant DCL4 is involved in the biogenesis of either functional endogenous or exogenous (i.e. viral) short interfering (si)RNAs, thus playing crucial antiviral roles.
Methods In this study we expressed plant DCL4 in Saccharomyces cerevisiae, an RNAi-depleted organism, in which we could highlight the role of dicing as neither Argonautes nor RNA-dependent RNA polymerase is present. We have therefore tested the DCL4 functionality in processing exogenous dsRNA-like substrates, such as a replicase-assisted viral replicon defective-interfering RNA and RNA hairpin substrates, or endogenous antisense transcripts.
Results DCL4 was shown to be functional in processing dsRNA-like molecules in vitro and in vivo into 21- and 22-nt sRNAs. Conversely, DCL4 did not efficiently process a replicase-assisted viral replicon in vivo, providing evidence that viral RNAs are not accessible to DCL4 in membranes associated in active replication. Worthy of note, in yeast cells expressing DCL4, 21- and 22-nt sRNAs are associated with endogenous loci.
Conclusions We provide new keys to interpret what was studied so far on antiviral DCL4 in the host system. The results all together confirm the role of sense/antisense RNA-based regulation of gene expression, expanding the sense/antisense atlas of S. cerevisiae. The results described herein show that S. cerevisiae can provide insights into the functionality of plant dicers and extend the S. cerevisiae tool to new biotechnological applications.
Keywords RNAi, Yeast, Plant viruses, DCL4, dsRNAs, Antisense transcript, Short RNAs
Address and Contact Information 1 Department of Biology, Agricultural and Food Sciences, National Research Council, Institute for Sustainable Plant Protection, Bari Unit, Bari, Italy
2 Department of Biomedical Sciences, National Research Council, Institute for Biomedical Technologies, Bari Unit, Bari, Italy
3 Institute of Biochemistry and Biotechnology, Section Microbial Biotechnology, Martin Luther University Halle-Wittenberg, Halle Saale, Germany
*Corresponding author: Vitantonio.pantaleo@cnr.it
Paola Leonetti, Arianna Consiglio and Dennis Arendt contributed equally to this work.
Read full article at BMC

No.  65DOI: 10.1186/s11658-023-00474-5 Volume 28 (2023) - 28:65
Title ASTROCYTE SENESCENCE-LIKE RESPONSE RELATED TO PERIPHERAL NERVE INJURY-INDUCED NEUROPATHIC PAIN
Authors Jingyi Du1†, Nan Cheng1†, Yifan Deng1, Ping Xiang2, Jianfen Liang1, Zhenye Zhang1, Ziqing Hei1 and Xiang Li1*
Abstract Background Peripheral nerve damage causes neuroinflammation, which plays a critical role in establishing and maintaining neuropathic pain (NeP). The mechanisms contributing to neuroinflammation remain poorly elucidated, and pharmacological strategies for NeP are limited. Thus, in this study, we planned to explore the possible link between astrocyte senescence and NeP disorders following chronic sciatic nerve injury.
Methods An NeP animal model was established by inducing chronic constrictive injury (CCI) to the sciatic nerve in adult rats. A senolytic drug combination of dasatinib and quercetin was gavaged daily from the first postoperative day until the end of the study. Paw mechanical withdrawal threshold (PMWT) and paw thermal withdrawal latency (PTWL) were evaluated to assess behaviors in response to pain in the experimental rats. Senescence-associated β-galactosidase staining, western blot analysis, and immunofluorescence were applied to examine the levels of proinflammatory factors and severity of the senescence-like response in the spinal cord. Lipopolysaccharide (LPS) was administered to induce senescence of spinal astrocytes in primary cultures in vitro, to explore the potential impacts of senescence on the secretion of proinflammatory factors. Furthermore, single-cell RNA sequencing (scRNA-seq) was conducted to identify senescence-related molecular responses in spinal astrocytes under neuropathic pain.
Results Following sciatic nerve CCI, rats exhibited reduced PMWT and PTWL, increased levels of spinal proinflammatory factors, and an enhanced degree of senescence in spinal astrocytes. Treatment with dasatinib and quercetin effectively attenuated spinal neuroinflammation and mitigated the hypersensitivities of the rats subjected to sciatic nerve CCI. Mechanistically, the dasatinib-quercetin combination reversed senescence in LPS-stimulated primary cultured astrocytes and decreased the levels of proinflammatory factors. The scRNA-seq data revealed four potential senescence-related genes in the spinal astrocyte population, and the expression of clusterin (CLU) protein was validated via in vitro experiments.
Conclusion The findings indicate the potential role of astrocyte senescence in neuroinflammation following peripheral nerve injury, and suggest that targeting CLU activation in astrocytes might provide a novel therapeutic strategy to treat NeP.
Keywords Neuropathic pain, Peripheral nerve injury, Neuroinfammation, Senescence, Astrocyte
Address and Contact Information 1 Department of Anesthesiology, The Third Affliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
2 Department of Medical Quality Management, Nanfang Hospital, Southern Medical University, Guangzhou 510000, China
*Corresponding author: lixiang27@mail.sysu.edu.cn
Jingyi Du and Nan Cheng contributed equally to this study.
Read full article at BMC

No.  66DOI: 10.1186/s11658-023-00482-5 Volume 28 (2023) - 28:66
Title MiR-15b-5p AND PCSK9 INHIBITION REDUCES LIPOPOLYSACCHARIDE-INDUCED ENDOTHELIAL DYSFUNCTION BY TARGETING SIRT4
Authors Elisa Martino1, Nunzia D’Onofrio1*, Anna Balestrieri2, Luigi Mele3, Celestino Sardu4, Rafaele Marfella4, Giuseppe Campanile5 and Maria Luisa Balestrieri1
Abstract Background Endothelial dysfunction and deregulated microRNAs (miRNAs) participate in the development of sepsis and are associated with septic organ failure and death. Here, we explored the role of miR-15b-5p on inflammatory pathways in lipopolysaccharide (LPS)-treated human endothelial cells, HUVEC and TeloHAEC.
Methods The miR-15b-5p levels were evaluated in LPS-stimulated HUVEC and TeloHAEC cells by quantitative real-time PCR (qRT–PCR). Functional experiments using cell counting kit-8 (CCK-8), transfection with antagomir, and enzyme-linked immunosorbent assays (ELISA) were conducted, along with investigation of pyroptosis, apoptosis, autophagy, and mitochondrial reactive oxygen species (ROS) by cytofluorometric analysis and verified by fluorescence microscopy. Sirtuin 4 (SIRT4) levels were detected by ELISA and immunoblotting, while proprotein convertase subtilisin-kexin type 9 (PCSK9) expression was determined by flow cytometry (FACS) and immunofluorescence analyses. Dual-luciferase reporter evaluation was performed to confirm the miR-15b-5p–SIRT4 interaction.
Results The results showed a correlation among miR-15b-5p, PCSK9, and SIRT4 levels in septic HUVEC and TeloHAEC. Inhibition of miR-15b-5p upregulated SIRT4 content, alleviated sepsis-related inflammatory pathways, attenuated mitochondrial stress, and prevented apoptosis, pyroptosis, and autophagic mechanisms. Finally, a PCSK9 inhibitor (i-PCSK9) was used to analyze the involvement of PCSK9 in septic endothelial injury. i-PCSK9 treatment increased SIRT4 protein levels, opposed the septic inflammatory cascade leading to pyroptosis and autophagy, and strengthened the protective role of miR-15b-5p inhibition. Increased luciferase signal validated the miR-15b-5p–SIRT4 binding.
Conclusions Our in vitro findings suggested the miR-15b-5p–SIRT4 axis as a suitable target for LPS-induced inflammatory pathways occurring in sepsis, and provide additional knowledge on the beneficial effect of i-PCSK9 in preventing vascular damage by targeting SIRT4.
Keywords microRNA-15b-5p, PCSK9, SIRT4, Sepsis, Endothelial infammation, Pyroptosis, Autophagy
Address and Contact Information 1 Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy
2 Food Safety Department, Istituto Zooproflattico Sperimentale del Mezzogiorno, Via Salute 2, 80055 Portici, Italy
3 Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Via Luciano Armanni 5, 80138 Naples, Italy
4 Department of Advanced Clinical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Miraglia, 80138 Naples, Italy
5 Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via F. Delpino 1, 80137 Naples, Italy
*Corresponding author: nunzia.donofrio@unicampania.it
Read full article at BMC

No.  67DOI: https://cmbl.biomedcentral.com/articles/10.1186/s11658-023-00480-7 Volume 28 (2023) - 28:67
Title PRENATAL LIPOPOLYSACCHARIDE EXPOSURE INDUCES ANXIETY-LIKE BEHAVIOUR IN MALE MOUSE OFFSPRING AND ABERRANT GLIAL DIFFERENTIATION OF EMBRYONIC NEURAL STEM CELLS
Authors Chie‐Pein Chen1,2, Pei‐Chun Chen2, Yu‐Ling Pan3 and Yi‐Chao Hsu3,4*
Abstract Background Prenatal infection has been implicated in the development of neuropsychiatric disorders in children. We hypothesised that exposure to lipopolysaccharide during prenatal development could induce anxiety-like behaviour and sensorineural hearing loss in offspring, as well as disrupt neural differentiation during embryonic neural development.
Methods We simulated prenatal infection in FVB mice and mouse embryonic stem cell (ESC) lines, specifically 46C and E14Tg2a, through lipopolysaccharide treatment. Gene expression profiling analyses and behavioural tests were utilized to study the effects of lipopolysaccharide on the offspring and alterations in toll-like receptor (TLR) 2-positive and TLR4-positive cells during neural differentiation in the ESCs.
Results Exposure to lipopolysaccharide (25 µg/kg) on gestation day 9 resulted in anxiety-like behaviour specifically in male offspring, while no effects were detected in female offspring. We also found significant increases in the expression of GFAP and CNPase, as well as higher numbers of GFAP + astrocytes and O4+ oligodendrocytes in the prefrontal cortex of male offspring. Furthermore, increased scores for genes related to oligodendrocyte and lipid metabolism, particularly ApoE, were observed in the prefrontal cortex regions. Upon exposure to lipopolysaccharide during the ESC-to-neural stem cell (NSC) transition, Tuj1, Map2, Gfap, O4, and Oligo2 mRNA levels increased in the differentiated neural cells on day 14. In vitro experiments demonstrated that lipopolysaccharide exposure induced inflammatory responses, as evidenced by increased expression of IL1b and ApoB mRNA.
Conclusions Our findings suggest that prenatal infection at different stages of neural differentiation may result in distinct disturbances in neural differentiation during ESC—NSC transitions. Furthermore, early prenatal challenges with lipopolysaccharide selectively induce anxiety-like behaviour in male offspring. This behaviour may be attributed to the abnormal differentiation of astrocytes and oligodendrocytes in the brain, potentially mediated by ApoB/E signalling pathways in response to inflammatory stimuli.
Keywords Lipopolysaccharide, Prenatal infection, Embryonic stem cell, Neural stem cell, Oligodendrocyte diferentiation, ApoB, ApoE
Address and Contact Information 1 Division of High Risk Pregnancy, Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
2 Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
3 Department of Audiology and Speech‐Language Pathology, MacKay Medical College, New Taipei City, Taiwan
4 Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
*Corresponding author: hsuyc@mmc.edu.tw
Read full article at BMC

No.  68DOI: 10.1186/s11658-023-00481-6 Volume 28 (2023) - 28:68
Title DESMOCOLLIN-1 IS ASSOCIATED WITH PRO-METASTATIC PHENOTYPE OF LUMINAL A BREAST CANCER CELLS AND IS MODULATED BY PARTHENOLIDE
Authors Petr Lapcik1, Petr Sulc1, Lucia Janacova1, Katerina Jilkova1, David Potesil2, Pavla Bouchalova1, Petr Müller3 and Pavel Bouchal1*
Abstract Background Desmocollin-1 (DSC1) is a desmosomal transmembrane glycoprotein that maintains cell-to-cell adhesion. DSC1 was previously associated with lymph node metastasis of luminal A breast tumors and was found to increase migration and invasion of MCF7 cells in vitro. Therefore, we focused on DSC1 role in cellular and molecular mechanisms in luminal A breast cancer and its possible therapeutic modulation.
Methods Western blotting was used to select potential inhibitor decreasing DSC1 protein level in MCF7 cell line. Using atomic force microscopy we evaluated effect of DSC1 overexpression and modulation on cell morphology. The LC–MS/MS analysis of total proteome on Orbitrap Lumos and RNA-Seq analysis of total transcriptome on Illumina NextSeq 500 were performed to study the molecular mechanisms associated with DSC1. Pull-down analysis with LC–MS/MS detection was carried out to uncover DSC1 protein interactome in MCF7 cells.
Results Analysis of DSC1 protein levels in response to selected inhibitors displays significant DSC1 downregulation (p-value ≤ 0.01) in MCF7 cells treated with NF-κB inhibitor parthenolide. Analysis of mechanic cell properties in response to DSC1 overexpression and parthenolide treatment using atomic force microscopy reveals that DSC1 overexpression reduces height of MCF7 cells and conversely, parthenolide decreases cell stiffness of MCF7 cells overexpressing DSC1. The LC–MS/MS total proteome analysis in data-independent acquisition mode shows a strong connection between DSC1 overexpression and increased levels of proteins LACRT and IGFBP5, increased expression of IGFBP5 is confirmed by RNA-Seq. Pathway analysis of proteomics data uncovers enrichment of proliferative MCM_BIOCARTA pathway including CDK2 and MCM2-7 after DSC1 overexpression. Parthenolide decreases expression of LACRT, IGFBP5 and MCM_BIOCARTA pathway specifically in DSC1 overexpressing cells. Pull-down assay identifies DSC1 interactions with cadherin family proteins including DSG2, CDH1, CDH3 and tyrosine kinase receptors HER2 and HER3; parthenolide modulates DSC1-HER3 interaction.
Conclusions Our systems biology data indicate that DSC1 is connected to mechanisms of cell cycle regulation in luminal A breast cancer cells, and can be effectively modulated by parthenolide.
Keywords DIA, Proteomics, Pull-down, DSC1, Breast cancer, Metastasis
Address and Contact Information 1 Department of Biochemistry, Faculty of Science, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
2 Central European Institute of Technology, Masaryk University, Brno, Czech Republic
3 Masaryk Memorial Cancer Institute, RECAMO, Brno, Czech Republic
*Corresponding author: bouchal@chemi.muni.cz
Read full article at BMC

No.  69DOI: 10.1186/s11658-023-00478-1 Volume 28 (2023) - 28:69
Title CORRECTION: INHIBITION OF CISD2 PROMOTES FERROPTOSIS THROUGH FERRITINOPHAGY-MEDIATED FERRITIN TURNOVER AND REGULATION OF p62–Keap1–NRF2 PATHWAY
Authors Yanchun Li1,2†>, Xueying Ren4†, Luyang Wang2, Yaqing Xu2, Yefeng Zhao2, Chen Yang2, Chen Yuan2, Huanjuan Li2, Xiangmin Tong2*, Ying Wang1* and Jing Du2*
Abstract Correction: Cellular & Molecular Biology Letters (2022) 27:81
https://doi.org/10.1186/s11658-022-00383-z Following publication of the original article [1], the author informed us that they recently received a remark from a reader who pointed out a discrepancy in Fig. 5a. Specifically, the reader noted that the background in the second column appears to be different from the background in the other columns. Therefore, we providing an explanation for this observed difference. The order of presentation for the xenografts was reorganized. The data pertaining to the group of shCISD2 with Keap1 overexpression was not included in the final manuscript. Therefore, the background in the second column appears to be different. The original figure is presented as Additional file 1. Neither of these changes affects the results and conclusions of this study. The authors extend their apologies for any inconvenience caused. Supplementary Information
The online version contains supplementary material available at https://doi.org/10.1186/s11658-023-00478-1.
Additional fle 1. The original figure of subcutaneous tumors in nude mice. Reference:
1. Li Y, Bing Xu, Ren X, Wang L, Yaqing Xu, Zhao Y, Yang C, Yuan C, Li H, Tong X, Wang Y, Jing Du. Inhibition of CISD2 pro-motes ferroptosis through ferritinophagy-mediated ferritin turnover and regulation of p62–Keap1–NRF2 pathway.
Cell Mol Biol Lett. 2022;27:81. https://doi.org/10.1186/s11658-022-00383-z.

Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional afliations.
Keywords
Address and Contact Information 1 Department of Central Laboratory, Afliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
2 Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People’s Hospital (Afliated People’s Hospital, Hangzhou Medical College), Hangzhou 310014, Zhejiang, China
3 Department of Clinical Laboratory, Hangzhou Women’s Hospital, Hangzhou 310016, Zhejiang, China
4 Department of Laboratory Medicine, The Second Affliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang, China
*Corresponding author: tongxiangmin@163.com; nancywangying@163.com; dujing1@hmc.edu.cn
Yanchun Li, Bing Xu and Xueying Ren contributed equally to this work.
The original article can be found online at https://doi.org/10.1186/s11658-022-00383-z.
Read full article at BMC

No.  70DOI: 10.1186/s11658-023-00477-2 Volume 28 (2023) - 28:70
Title CRISPR/Cas13d TARGETING GZMA IN PARs PATHWAY REGULATES THE FUNCTION OF OSTEOCLASTS IN CHRONIC APICAL PERIODONTITIS
Authors Tingting Jia1, Fang Yuan2, Jingqiao Tao3, Gang Wang4, Xianhua Zhang1, Bin Zhang1* and Hongbo Li1*
Abstract Chronic apical periodontitis is a prevalent oral disease characterized by bone loss, and its underlying mechanisms remain unclear. This study aimed to investigate the role and mechanism of the serine protease GZMA in osteoclasts during chronic apical periodontitis. To address this, we employed crRNA/Cas13d to inhibit GZMA expression and examined its impact on osteoclast behavior. Our findings revealed that GZMA plays a significant role in promoting osteoclast cell proliferation while inhibiting cell apoptosis. Additionally, the inhibition of GZMA led to a notable increase in miR-25-3p expression, which, in turn, downregulated the expression of TGF-β. Consequently, the reduction in TGF-β expression led to a decrease in PAR1 expression within the PARs pathway. These results suggest that GZMA might serve as a promising therapeutic target for the treatment of chronic apical periodontitis. Furthermore, our study highlights the potential of targeting GZMA using crRNA/Cas13d as a valuable approach for future therapeutic interventions.
Keywords GZMA, CRISPR/Cas13d, Osteoclast, Chronic apical periodontitis
Address and Contact Information 1 Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
2 Department of Oncology, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
3 Department of Stomatology, Southern Medical Branch of PLA General Hospital, Beijing, China
4 Medical School of Chinese PLA, Beijing, China
*Corresponding author: binzhangfmmu@163.com; hongbolii@sina.com
Read full article at BMC

No.  71DOI: 10.1186/s11658-023-00484-3 Volume 28 (2023) - 28:71
Title UGCG MODULATES HEART HYPERTROPHY THROUGH B4GalT5-MEDIATED MITOCHONDRIAL OXIDATIVE STRESS AND THE ERK SIGNALING PATHWAY
Authors Shengyu Cui1,2,3†, Xutao Zhang1,2,3†, Yuhua Li4, Shan Hu1,2,3, Bing Wu1,2,3, Zhao Fang1,2,3, Jixian Gao1,2,3, Ming Li1,2,3, Haoliang Wu1,2,3, Bo Tao1,2,3, Hao Xia1,2,3* and Lin Xu5*
Abstract Mechanical pressure overload and other stimuli often contribute to heart hypertrophy, a significant factor in the induction of heart failure. The UDP-glucose ceramide glycosyltransferase (UGCG) enzyme plays a crucial role in the metabolism of sphingolipids through the production of glucosylceramide. However, its role in heart hypertrophy remains unknown. In this study, UGCG was induced in response to pressure overload in vivo and phenylephrine stimulation in vitro. Additionally, UGCG downregulation ameliorated cardiomyocyte hypertrophy, improved cardiomyocyte mitochondrial oxidative stress, and reduced the ERK signaling pathway. Conversely, UGCG overexpression in cardiomyocytes promoted heart hypertrophy development, aggravated mitochondrial oxidative stress, and stimulated ERK signaling. Furthermore, the interaction between beta-1,4-galactosyltransferase 5 (B4GalT5), which catalyses the synthesis of lactosylceramide, and UGCG was identified, which also functions as a synergistic molecule of UGCG. Notably, limiting the expression of B4GalT5 impaired the capacity of UGCG to promote myocardial hypertrophy, suggesting that B4GalT5 acts as an intermediary for UGCG. Overall, this study highlights the potential of UGCG as a modulator of heart hypertrophy, rendering it a potential target for combating heart hypertrophy.
Keywords Heart hypertrophy, UGCG, B4GalT5, ERK signaling
Address and Contact Information 1 Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
2 Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
3 Hubei Key Laboratory of Cardiology, Wuhan 430060, China
4 Intensive Care Unit, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
5 Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China
*Correspondence: xiahao1966@163.com; linxu2018@whu.edu.cn
Shengyu Cui and Xutao Zhang are co-first authors and equal contributed to this work.
Read full article at BMC

No.  72DOI: 10.1186/s11658-023-00475-4 Volume 28 (2023) - 28:72
Title N6-METHYLADENOSINE MODIFICATION OF PLOD2 CAUSES SPERMATOCYTE DAMAGE IN RATS WITH VARICOCELE
Authors Huan Li1†, Jun Zhao1†, Hao Deng1†, YuCheng Zhong1†, Mian Chen2†, LinSheng Chi1, GuoQun Luo1, Cong Cao1, Cong Yu3, Honghai Liu3 and Xinzong Zhang4*
Abstract Background: In recent years, N6-methyladenosine (m6A) methylation modification of mRNA has been studied extensively. It has been reported that m6A determines mRNA fate and participates in many cellular functions and reactions, including oxidative stress. The PLOD2 gene encodes a protein that plays a key role in tissue remodeling and fibrotic processes.
Methods: The m6A methylation and expression levels of PLOD2 were determined by m6A methylated RNA immunoprecipitation sequencing (MeRIP-seq) and MeRIP-quantitative polymerase chain reaction (qPCR) in the testes of varicocele rats compared with control. To determine whether IGF2BP2 had a targeted effect on the PLOD2 mRNA, RNA immunoprecipitation-qPCR (RIP-qPCR) and luciferase assays were performed. CRISPR/dCas13b-ALKBH5 could downregulate m6A methylation level of PLOD2, which plays an important role in PLOD2-mediated cell proliferation and apoptosis in GC-2 cells.
Results: PLOD2 was frequently exhibited with high m6A methylation and expression level in the testes of varicocele rats compared with control. In addition, we found that IGF2BP2 binds to the m6A-modified 3′ untranslated region (3′-UTR) of PLOD2 mRNA, thereby positively regulating its mRNA stability. Targeted specific demethylation of PLOD2 m6A by CRISPR/dCas13b-ALKBH5 system can significantly decrease the m6A and expression level of PLOD2. Furthermore, demethylation of PLOD2 mRNA dramatically promote GC-2 cell proliferation and inhibit cell apoptosis under oxidative stress.
Conclusion: As a result, we found that varicocele-induced oxidative stress promoted PLOD2 expression level via m6A methylation modification. In addition, targeting m6A demethylation of PLOD2 by CRISPR/dCas13b-ALKBH5 system can regulate GC-2 cell proliferation and apoptosis under oxidative stress. Taken together, our study has acquired a better understanding of the mechanisms underlying male infertility associated with oxidative stress, as well as a novel therapeutic target for male infertility.
Keywords PLOD2, m6A, Varicocele, GC-2 cell, Rat
Address and Contact Information 1 Assisted Reproductive Technology Center, Foshan Maternal and Child Health Care Hospital, Foshan, China
2 Pharmacy Department, Foshan Maternal and Child Health Care Hospital, Foshan, China
3 Assisted Reproductive Technology Centre, Maternity and Child Healthcare Hospital of Meizhou, Meizhou, China
4 NHC Key Laboratory of Male Reproduction and Genetics, Guangdong Provincial Reproductive Science Institute (Guangdong Provincial Fertility Hospital), Guangzhou, China
*Corresponding author: 13857170787@139.com
Huan Li, Jun Zhao, Hao Deng, YuCheng Zhong and Mian Chen contributed equally to this work.
Read full article at BMC

No.  73DOI: 10.1186/s11658-023-00483-4 Volume 28 (2023) - 28:73
Title CRISPR APPLICATIONS IN CANCER DIAGNOSIS AND TREATMENT
Authors Mingxia Wang1,2†, Menghui Chen1†, Xia Wu1, Xinbo Huang1* and Bo Yu1*
Abstract Cancer remains a significant global health challenge, necessitating the exploration of novel and more precise therapeutic options beyond conventional treatments. In this regard, clustered regularly interspaced short palindromic repeats (CRISPR) systems have emerged as highly promising tools for clinical gene editing applications. The CRISPR family encompasses diverse CRISPR-associated (Cas) proteins that possess the ability to recognize specific target sequences. The initial CRISPR system consisted of the Cas9 protein and a single-guide RNA, which guide Cas9 to the desired target sequence, facilitating precise double-stranded cleavage. In addition to the traditional cis-cleavage activity, the more recently discovered Cas12 and Cas13 proteins exhibit trans-cleavage activity, which expands their potential applications in cancer diagnosis. In this review, we provide an overview of the functional characteristics of Cas9, Cas12, and Cas13. Furthermore, we highlight the latest advancements and applications of these CRISPR systems in cancer gene therapy and molecular diagnosis. We also emphasize the importance of understanding the strengths and limitations of each CRISPR system to maximize their clinical utility. By providing a comprehensive overview of the current state of CRISPR technology in cancer research, we aim to inspire further exploration and innovation in this rapidly evolving field.
Keywords Cas9, Cas12, Cas13, Cancer, Gene therapy, Diagnostic tools, Clinical trials
Address and Contact Information 1 Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen 518036, China
2 Shenzhen Key Laboratory of Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen 518000, China
* Corresponding author: huangxinbo92@pku.edu.cn; drboyu_derm@126.com
Mingxia Wang and Menghui Chen contributed equally to this work and considered as co-first authors.
Read full article at BMC

No.  74DOI: 10.1186/s11658-023-00486-1 Volume 28 (2023) - 28:74
Title S100a9 INHIBITS Atg9a TRANSCRIPTION AND PARTICIPATES IN SUPPRESSION OF AUTOPHAGY IN CARDIOMYOCYTES INDUCED BY β1-ADRENOCEPTOR AUTOANTIBODIES
Authors Xiaoyan Zhi1, Shu Shi1, Yang Li1, Mingxia Ma1, Yaolin Long1, Chen Li1, Haihu Hao2, Huirong Liu3, Xiaohui Wang1 and Li Wang1*
Abstract Background: Cardiomyocyte death induced by autophagy inhibition is an important cause of cardiac dysfunction. In-depth exploration of its mechanism may help to improve cardiac dysfunction. In our previous study, we found that β1-adrenergic receptor autoantibodies (β1-AAs) induced a decrease in myocardial autophagy and caused cardiomyocyte death, thus resulting in cardiac dysfunction. Through tandem mass tag (TMT)-based quantitative proteomics, autophagy-related S100a9 protein was found to be significantly upregulated in the myocardial tissue of actively immunized mice. However, whether S100a9 affects the cardiac function in the presence of β1-AAs through autophagy and the specific mechanism are currently unclear.
Methods: In this study, the active immunity method was used to establish a β1-AA-induced mouse cardiac dysfunction model, and RT-PCR and western blot were used to detect changes in gene and protein expression in cardiomyocytes. We used siRNA to knockdown S100a9 in cardiomyocytes. An autophagy PCR array was performed to screen differentially expressed autophagy-related genes in cells transfected with S100a9 siRNA and negative control siRNA. Cytoplasmic nuclear separation, co-immunoprecipitation (Co-IP), and immunofluorescence were used to detect the binding of S100a9 and hypoxia inducible factor-1α (HIF-1α). Finally, AAV9-S100a9-RNAi was injected into mice via the tail vein to knockdown S100a9 in cardiomyocytes. Cardiac function was detected via ultrasonography.
Results: The results showed that β1-AAs induced S100a9 expression. The PCR array indicated that Atg9a changed significantly in S100a9siRNA cells and that β1-AAs increased the binding of S100a9 and HIF-1α in cytoplasm. Knockdown of S100a9 significantly improved autophagy levels and cardiac dysfunction.
Conclusion: Our research showed that β1-AAs increased S100a9 expression in cardiomyocytes and that S100a9 interacted with HIF-1α, which prevented HIF-1α from entering the nucleus normally, thus inhibiting the transcription of Atg9a. This resulted in autophagy inhibition and cardiac dysfunction.
Keywords S100a9, βS100a9, β1-AA, Autophagy, HIF-1α, Atg9a-AA, Autophagy, HIF-1α, Atg9a
Address and Contact Information 1 Department of Pathology, Shanxi Medical University, No.56 Xinjian South Road, Taiyuan, Shanxi 030001, People’s Republic of China
2 Department of Orthopaedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
3 Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
*Corresponding author: mirror0117@126.com
Read full article at BMC

No.  75DOI: 10.1186/s11658-023-00485-2 Volume 28 (2023) - 28:75
Title ENHANCEMENT OF THE THERAPEUTIC EFFICACY OF MESENCHYMAL STEM CELL-DERIVED EXOSOMES IN OSTEOARTHRITIS
Authors Zehao Zhang1, Sheng Zhao1, Zhaofeng Sun1, Chuanxing Zhai1, Jiang Xia2, Caining Wen3*, Yuge Zhang3* and Yuanmin Zhang3*
Abstract Osteoarthritis (OA), a common joint disorder with articular cartilage degradation as the main pathological change, is the major source of pain and disability worldwide. Despite current treatments, the overall treatment outcome is unsatisfactory. Thus, patients with severe OA often require joint replacement surgery. In recent years, mesenchymal stem cells (MSCs) have emerged as a promising therapeutic option for preclinical and clinical palliation of OA. MSC-derived exosomes (MSC-Exos) carrying bioactive molecules of the parental cells, including non-coding RNAs (ncRNAs) and proteins, have demonstrated a significant impact on the modulation of various physiological behaviors of cells in the joint cavity, making them promising candidates for cell-free therapy for OA. This review provides a comprehensive overview of the biosynthesis and composition of MSC-Exos and their mechanisms of action in OA. We also discussed the potential of MSC-Exos as a therapeutic tool for modulating intercellular communication in OA. Additionally, we explored bioengineering approaches to enhance MSC-Exos’ therapeutic potential, which may help to overcome challenges and achieve clinically meaningful OA therapies.
Keywords Osteoarthritis, Mesenchymal stem cells, Exosomes, Bioengineering
Address and Contact Information 1 School of Clinical Medicine, Jining Medical University, Jining 272067, Shandong, China
2 Department of Chemistry, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
3 Department of Joint Surgery and Sports Medicine, Afliated Hospital of Jining Medical University, Jining Medical University, Jining 272029, Shandong, China
*Corresponding author: 547645878@qq.com; 1989114716@qq.com; yuanminzhang@mail.jnmc.edu.cn
Read full article at BMC

No.  76DOI: 10.1186/s11658-023-00489-y Volume 28 (2023) - 28:76
Title OXIDATIVE STRESS AS A KEY MODULATOR OF CELL FATE DECISION IN OSTEOARTHRITIS AND OSTEOPOROSIS: A NARRATIVE REVIEW
Authors Jana Riegger1*, Astrid Schoppa2, Leonie Ruths1, Melanie Hafner‐Luntzer2 and Anita Ignatius2
Abstract During aging and after traumatic injuries, cartilage and bone cells are exposed to various pathophysiologic mediators, including reactive oxygen species (ROS), damage-associated molecular patterns, and proinflammatory cytokines. This detrimental environment triggers cellular stress and subsequent dysfunction, which not only contributes to the development of associated diseases, that is, osteoporosis and osteoarthritis, but also impairs regenerative processes. To counter ROS-mediated stress and reduce the overall tissue damage, cells possess diverse defense mechanisms. However, cellular antioxidative capacities are limited and thus ROS accumulation can lead to aberrant cell fate decisions, which have adverse effects on cartilage and bone homeostasis. In this narrative review, we address oxidative stress as a major driver of pathophysiologic processes in cartilage and bone, including senescence, misdirected differentiation, cell death, mitochondrial dysfunction, and impaired mitophagy by illustrating the consequences on tissue homeostasis and regeneration. Moreover, we elaborate cellular defense mechanisms, with a particular focus on oxidative stress response and mitophagy, and briefly discuss respective therapeutic strategies to improve cell and tissue protection.
Keywords Osteoarthritis, Osteoporosis, Senescence, Oxidative stress, ROS, Cell death, Cell fate decision, Mitochondrial dysfunction, Cartilage, Bone
Address and Contact Information 1 Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081 Ulm, Germany
2 Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
* Corresponding author: jana.riegger@uni-ulm.de
Read full article at BMC

No.  77DOI: 10.1186/s11658-023-00492-3 Volume 28 (2023) - 28:77
Title lncRNA Helf pROMOTES HEPATIC INFLAMMATION AND FIBROSIS BY INTERACTING WITH PTBP1 TO FACILITATE PIK3R5 mRNA STABILIZATION
Authors Xiaohui Han1†, Beichen Guo2†, Sicong Zhao1†, Yehua Li1, Jing Zhu1, Yifan He1, Jiajun Wang1, Qingbin Yao1, Shuai Shao2, Lina Zheng1, Zhemin Shi1, Tao Han2, Wei Hong1 and Kun Zhang1*
Abstract Background: Hepatic fibrosis is a common consequence of chronic liver diseases without approved antifibrotic therapies. Long noncoding RNAs (lncRNAs) play an important role in various pathophysiological processes. However, the functions of certain lncRNAs involved in mediating the antifibrotic role remain largely unclear.
Methods: The RNA level of lnc-High Expressed in Liver Fibrosis (Helf) was detected in both mouse and human fibrotic livers. Furthermore, lnc-Helf-silenced mice were treated with carbon tetrachloride (CCl4) or bile duct ligation (BDL) to investigate the function of lnc-Helf in liver fibrosis.
Results: We found that lnc-Helf has significantly higher expression in human and mouse fibrotic livers as well as M1 polarized hepatic macrophages (HMs) and activated hepatic stellate cells (HSCs). In vivo studies showed that silencing lnc-Helf by AAV8 vector alleviates CCl4- and BDL-induced hepatic inflammation and fibrosis. Furthermore, in vitro experiments revealed that lnc-Helf promotes HSCs activation and proliferation, as well as HMs M1 polarization and proliferation in the absence or presence of cytokine stimulation. Mechanistically, our data illustrated that lnc-Helf interacts with RNA binding protein PTBP1 to promote its interaction with PIK3R5 mRNA, resulting in increased stability and activating the AKT pathway, thus promoting HSCs and HMs activation and proliferation, which augments hepatic inflammation and fibrosis.
Conclusion: Our results unveil a lnc-Helf/PTBP1/PIK3R5/AKT feedforward, amplifying signaling that exacerbates the process of hepatic inflammation and fibrosis, thus providing a possible therapeutic strategy for hepatic fibrosis.
Keywords
Address and Contact Information 1 Department of Histology and Embryology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin 300070, China
2 Department of Hepatology and Gastroenterology, Tianjin Union Medical Center Afliated to Nankai University, Tianjin, China
*Corresponding author: zhangkun@tmu.edu.cn
Xiaohui Han, Beichen Guo and Sicong Zhao contributed equally.
Read full article at BMC

No.  78DOI: 10.1186/s11658-023-00490-5 Volume 28 (2023) - 28:78
Title N6-METHYLADENOSINE MODIFICATION—A KEY PLAYER IN VIRAL INFECTION
Authors Xiaoyue Zhang1,4, Qiu Peng1* and Lujuan Wang2,3*
Abstract N6
-methyladenosine (m6A) modification is a dynamic, reversible process and is the most prevalent internal modification of RNA. This modification is regulated by three protein groups: methyltransferases (“writers”), demethylases (“erasers”), and m6A-binding proteins (“readers”). m6A modification and related enzymes could represent an optimal strategy to deepen the epigenetic mechanism. Numerous reports have suggested that aberrant modifications of m6A lead to aberrant expression of important viral genes. Here, we review the role of m6A modifications in viral replication and virus–host interactions. In particular, we focus on DNA and RNA viruses associated with human diseases, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), human immunodeficiency virus (HIV)-1, Epstein–Barr virus (EBV), and Kaposi’s sarcoma-associated herpesvirus (KSHV). These findings will contribute to the understanding of the mechanisms of virus–host interactions and the design of future therapeutic targets for treatment of tumors associated with viral infections.
Keywords m6A modifcation, DNA virus, RNA virus, Viral infection
Address and Contact Information 1 Hunan Cancer Hospital and the Afiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Changsha 410013, Hunan, China
2 Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
3 Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
4 Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
* Corresponding author: pengqiu@hnca.org.cn; wanglujuan001@csu.edu.cn
Read full article at BMC

No.  79DOI: 10.1186/s11658-023-00494-1 Volume 28 (2023) - 28:79
Title ACTIVATION OF BZW1 BY CEBPB IN MACROPHAGES PROMOTES eIF2α PHOSPHORYLATION-MEDIATED METABOLIC REPROGRAMMING AND ENDOPLASMIC RETICULUM STRESS IN MRL/lpr LUPUS-PRONE MICE
Authors Huimeng Qi1†, Zhaoguo Zheng2† and Qiang Liu*
Abstract Background: Lupus nephritis (LN) is associated with significant mortality and morbidity, while effective therapeutics and biomarkers are limited since the pathogenesis is complex. This study investigated the roles of the CEBPB/BZW1/eIF2α axis in metabolic reprogramming and endoplasmic reticulum stress in LN.
Method: The differentially expressed genes in LN were screened using bioinformatics tools. The expression of CEBPB in the renal tissue of patients with LN and its correlation with the levels of creatinine and urinary protein were analyzed. We used adenoviral vectors to construct LN mice with knockdown CEBPB using MRL/lpr lupus-prone mice and analyzed the physiological and autoimmune indices in mice. Chromatin immunoprecipitation quantitative polymerase chain reaction (ChIP–qPCR) and dual-luciferase reporter assays were conducted to explore the regulation of BZW1 by CEBPB, followed by glycolytic flux analysis, glucose uptake, and enzyme-linked immunosorbent assay (ELISA). Finally, the role of eIF2α phosphorylation by BZW1 in bone marrow-derived macrophages (BMDM) was explored using eIF2α phosphorylation and endoplasmic reticulum stress inhibitors.
Results: CEBPB was significantly increased in renal tissues of patients with LN and positively correlated with creatinine and urine protein levels in patients. Downregulation of CEBPB alleviated the autoimmune response and the development of nephritis in LN mice. Transcriptional activation of BZW1 by CEBPB-mediated glucose metabolic reprogramming in macrophages, and upregulation of BZW1 reversed the mitigating effect of CEBPB knockdown on LN. Regulation of eIF2α phosphorylation levels by BZW1 promoted endoplasmic reticulum stress-amplified inflammatory responses in BMDM.
Conclusion: Transcriptional activation of BZW1 by CEBPB promoted phosphorylation of eIF2α to promote macrophage glycolysis and endoplasmic reticulum stress in the development of LN.
Keywords CEBPB, BZW1, Glycolysis, Endoplasmic reticulum stress, Lupus nephritis
Address and Contact Information 1 Department of General Practice, Fuyang Hospital of Anhui Medical University, Fuyang 236000, Anhui, People’s Republic of China
2 Department of Nephrology, Guangdong Second Provincial General Hospital, Haizhu District, No. 466, Xingang Zhong, Guangzhou 510317, Guangdong, People’s Republic of China
*Corresponding author: liuqiang@ahmu.edu.cn
Huimeng Qi and Zhaoguo Zheng contributed equally to this work.
Read full article at BMC

No.  80DOI: 10.1186/s11658-023-00487-0 Volume 28 (2023) - 28:80
Title MCCC2 IS A NOVEL MEDIATOR BETWEEN MITOCHONDRIA AND TELOMERE AND FUNCTIONS AS AN ONCOGENE IN COLORECTAL CANCER
Authors Wanjun Liu1,2†, Si Chen1†, Wenqing Xie1,2, Qian Wang3, Qianxin Luo3, Minghan Huang1,2, Minyi Gu2,4, Ping Lan1,5* and Daici Chen1,2*
Abstract Background: The mitochondrial gene MCCC2, a subunit of the heterodimer of 3-methylcrotonyl-CoA carboxylase, plays a pivotal role in catabolism of leucine and isovaleric acid. The molecular mechanisms and prognostic value still need to be explored in the context of specific cancers, including colorectal cancer (CRC).
Methods: In vitro and in vivo cell-based assays were performed to explore the role of MCCC2 in CRC cell proliferation, invasion, and migration. Mitochondrial morphology, membrane potential, intracellular reactive oxygen species (ROS), telomerase activity, and telomere length were examined and analyzed accordingly. Protein complex formation was detected by co-immunoprecipitation (CO-IP). Mitochondrial morphology was observed by transmission electron microscopy (TEM). The Cancer Genome Atlas (TCGA) CRC cohort analysis, qRT-PCR, and immunohistochemistry (IHC) were used to examine the MCCC2 expression level. The association between MCCC2 expression and various clinical characteristics was analyzed by chi-square tests. CRC patients’ overall survival (OS) was analyzed by Kaplan–Meier analysis.
Results: Ectopic overexpression of MCCC2 promoted cell proliferation, invasion, and migration, while MCCC2 knockdown (KD) or knockout (KO) inhibited cell proliferation, invasion, and migration. MCCC2 KD or KO resulted in reduced mitochondria numbers, but did not affect the gross ATP production in the cells. Mitochondrial fusion markers MFN1, MFN2, and OPA1 were all upregulated in MCCC2 KD or KO cells, which is in line with a phenomenon of more prominent mitochondrial fusion. Interestingly, telomere lengths of MCCC2 KD or KO cells were reduced more than control cells. Furthermore, we found that MCCC2 could specifically form a complex with telomere binding protein TRF2, and MCCC2 KD or KO did not affect the expression or activity of telomerase reverse transcriptase (TERT). Finally, MCCC2 expression was heightened in CRC, and patients with higher MCCC2 expression had favorable prognosis.
Conclusions: Together, we identified MCCC2 as a novel mediator between mitochondria and telomeres, and provided an additional biomarker for CRC stratification.
Keywords MCCC2, Mitochondria, Telomere, Colorectal cancer
Address and Contact Information 1 Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Afiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou 510655, Guangdong, China
2 Biomedical Innovation Center, The Sixth Afiliated Hospital, Sun Yat-sen University, Guangzhou, China
3 Department of Intensive Care Unit, The Sixth Afiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou 510655, China
4 Scientifc Journal Center, The Sixth Afiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou 510655, China
5 Department of General Surgery, The Sixth Afiliated Hospital, Sun Yat-sen University, 26 Yuancun Er Heng Road, Guangzhou 510655, China
*Corresponding author: lanping@mail.sysu.edu.cn; chendc3@mail.sysu.edu.cn
Wanjun Liu, Si Chen and Wenqing Xie contributed equally to this work.
Read full article at BMC

No.  81DOI: 10.1186/s11658-023-00491-4 Volume 28 (2023) - 28:81
Title MACROPHAGE-DRIVEN CARDIAC INFLAMMATION AND HEALING: INSIGHTS FROM HOMEOSTASIS AND MYOCARDIAL INFARCTION
Authors Wenjie Zuo1†, Renhua Sun2†, Zhenjun Ji1 and Genshan Ma1*
Abstract Early and prompt reperfusion therapy has markedly improved the survival rates among patients enduring myocardial infarction (MI). Nonetheless, the resulting adverse remodeling and the subsequent onset of heart failure remain formidable clinical management challenges and represent a primary cause of disability in MI patients worldwide. Macrophages play a crucial role in immune system regulation and wield a profound influence over the inflammatory repair process following MI, thereby dictating the degree of myocardial injury and the subsequent pathological remodeling. Despite numerous previous biological studies that established the classical polarization model for macrophages, classifying them as either M1 pro-inflammatory or M2 pro-reparative macrophages, this simplistic categorization falls short of meeting the precision medicine standards, hindering the translational advancement of clinical research. Recently, advances in single-cell sequencing technology have facilitated a more profound exploration of macrophage heterogeneity and plasticity, opening avenues for the development of targeted interventions to address macrophage-related factors in the aftermath of MI. In this review, we provide a summary of macrophage origins, tissue distribution, classification, and surface markers. Furthermore, we delve into the multifaceted roles of macrophages in maintaining cardiac homeostasis and regulating inflammation during the post-MI period.
Keywords Macrophage, Myocardial infarction, Infammation, Polarization, Hemostasis
Address and Contact Information 1 Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, No. 87, Dingjiaqiao, Nanjing 210009, China
2 Department of Cardiology, Yancheng No. 1 People’s Hospital, No. 66 South Renmin Road, Yancheng 224000, China
*Corresponding author: magenshan@hotmail.com
Wenjie Zuo and Renhua Sun have contributed equally to this work and share first authorship.
Read full article at BMC

No.  82DOI: 10.1186/s11658-023-00502-4 Volume 28 (2023) - 28:82
Title CORRECTION: PROFILING THE IMPACT OF THE PROMOTERS ON CRISPR-Cas12a SYSTEM IN HUMAN CELLS
Authors Jinhe Li2†, Qingchun Liang2,3†, HuaPing Zhou1, Ming Zhou1* and Hongxin Huang1*
Abstract The Original Article was published on 17 May 2023

Correction: Cellular & Molecular Biology Letters (2023) 28:41 https://doi.org/10.1186/s11658-023-00454-9

Following publication of the original article [1], we have been informed that the name of author Qingchun Liang has a spelling.

The incorrect name is: Qinchun Liang
The correct name is: Qingchun Liang
Reference
1. Li J, Liang Q, Zhou HP, Zhou M, Huang H. Profiling the impact of the promoters on CRISPR-Cas12a system in human cells. Cell Mol Biol Lett. 2023;28:41. https://doi.org/10.1186/s11658-023-00454-9.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affliations.
Keywords
Address and Contact Information 1 Afiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China
2 The Third Afiliated Hospital of Southern Medical University, Guangzhou 510630, China
3 Guangzhou Key Laboratory of Neuropathic Pain Mechanism at Spinal Cord Level, Guangzhou 510630, China
*Corresponding author: zhouming@gzhmu.edu.cn; huanghx08@gzhmu.edu.cn † Jinhe Li and Qingchun Liang equally contributed to this work.
The original article can be found online at https://doi.org/10.1186/s11658-023-00454-9.
Read full article at BMC

No.  83DOI: 10.1186/s11658-023-00496-z Volume 28 (2023) - 28:83
Title A CONSERVED ZFX/WNT3 AXIS MODULATES THE GROWTH AND IMATINIB RESPONSE OF CHRONIC MYELOID LEUKEMIA STEM/PROGENITOR CELLS
Authors Xiuyan Zhang1,2*, Yu Wang1 , Jinchang Lu1 , Lun Xiao3 , Hui Chen1 , Quanxue Li4 , Yuan‐Yuan Li4 , Peng Xu1 , Changgeng Ruan2,5,6,7, Haixia Zhou2,5,7* and Yun Zhao1,5,6,7*
Abstract Background: Zinc finger protein X-linked (ZFX) has been shown to promote the growth of tumor cells, including leukemic cells. However, the role of ZFX in the growth and drug response of chronic myeloid leukemia (CML) stem/progenitor cells remains unclear.
Methods: Real-time quantitative PCR (RT–qPCR) and immunofluorescence were used to analyze the expression of ZFX and WNT3 in CML CD34+ cells compared with normal control cells. Short hairpin RNAs (shRNAs) and clustered regularly interspaced short palindromic repeats/dead CRISPR-associated protein 9 (CRISPR/dCas9) technologies were used to study the role of ZFX in growth and drug response of CML cells. Microarray data were generated to compare ZFX-silenced CML CD34+ cells with their controls. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays were performed to study the molecular mechanisms of ZFX to regulate WNT3 expression. RT–qPCR and western blotting were used to study the effect of ZFX on β-catenin signaling.
Results: We showed that ZFX expression was significantly higher in CML CD34+ cells than in control cells. Overexpression and gene silencing experiments indicated that ZFX promoted the in vitro growth of CML cells, conferred imatinib mesylate (IM) resistance to these cells, and enhanced BCR/ABL-induced malignant transformation. Microarray data and subsequent validation revealed that WNT3 transcription was conservatively regulated by ZFX. WNT3 was highly expressed in CML CD34+ cells, and WNT3 regulated the growth and IM response of these cells similarly to ZFX. Moreover, WNT3 overexpression partially rescued ZFX silencing-induced growth inhibition and IM hypersensitivity. ZFX silencing decreased WNT3/β-catenin signaling, including c-MYC and CCND1 expression.
Conclusion: The present study identified a novel ZFX/WNT3 axis that modulates the growth and IM response of CML stem/progenitor cells.
Keywords ZFX, WNT3, CD34+ cells, Chronic myeloid leukemia, Imatinib mesylate
Address and Contact Information 1 Cyrus Tang Medical Institute, Soochow University, Suzhou 215123, China
2 Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, The First Afiliated Hospital of Soochow University, Suzhou 215006, China
3 Department of Vascular Surgery, The Afiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
4 Shanghai‐MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
5 National Clinical Research Center for Hematologic Diseases, Suzhou 215006, China
6 Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China
7 MOE Engineering Center of Hematological Disease, Soochow University, Suzhou 21513, China
* Correspondig author: zhangxiuyan@suda.edu. cn; zhouhaixia@suda.edu.cn; zhaoy@suda.edu.cn
Read full article at BMC

No.  84DOI: 10.1186/s11658-023-00499-w Volume 28 (2023) - 28:84
Title RETRACTION NOTE: MicroRNA-325-3p INHIBITS CELL PROLIFERATION AND INDUCES APOPTOSIS IN HEPATITIS B VIRUSRELATED HEPATOCELLULAR CARCINOMA BY DOWNREGULATION OF AQUAPORIN 5
Authors Zhitao Zhang2 , Yanzhen Han1* , Guangxin Sun1 , Xiaohong Liu1 , Xiaoyan Jia1 and Xiangjun Yu1
Abstract The Original Article was published on 12 February 2019

Retraction Note : Cellular & Molecular Biology Letters (2019) 24:13 https://doi.org/10.1186/s11658-019-0137-1

The Editor in Chief has retracted this article because of concerns about the data presented. Specifically, the flow cytometry plots presented in Fig. 1E show a significant similarity to the plots presented in a previously published paper with no common authors [1]. Additionally, the ACP5 blots presented in Fig. 2F appear highly similar to two blots presented in a paper published around the same time as this paper with no common authors [2]. The Editor in Chief no longer has confidence in the reliability of the data. All authors agree to this retraction.
References
1. Tian R, Li Y, Gao M. Shikonin causes cell-cycle arrest and induces apoptosis by regulating the EGFR-NF-κB signalling pathway in human epidermoid carcinoma A431 cells. Biosci Rep. 2015;35(2): e00189. https://doi.org/10.1042/BSR20150002.
2. Chen H, Zhang Y, Su H, Shi H, Xiong Q, Su Z. Overexpression of miR-1283 inhibits cell proliferation and invasion of glioma cells by targeting ATF4. Oncol Res. 2019;27(3):325–34. https://doi.org/10.3727/096504018X15251282086836.

Keywords
Address and Contact Information 1 General Surgery V Ward, Afliated Hospital of Hebei Engineering University, Handan 056002, Hebei, People’s Republic of China
2 Clinical Laboratory, Handan Infectious Disease Hospital, Handan 056002, Hebei, People’s Republic of China
*Corresponding author: hanyanzhenhebei@163.com
The original article can be found online at https://doi.org/10.1186/s11658-019-0137-1.
Read full article at BMC

No.  85DOI: 10.1186/s11658-023-00500-6 Volume 28 (2023) - 28:85
Title INHIBITION OF SQSTM1 S403 PHOSPHORYLATION FACILITATES THE AGGRESOME FORMATION OF UBIQUITINATED PROTEINS DURING PROTEASOME DYSFUNCTION
Authors Chenliang Zhang1*, YiChun Duan2, Chen Huang2 and Liping Li3
Abstract Background: Ubiquitin–proteasome-system-mediated clearance of misfolded proteins is essential for cells to maintain proteostasis and reduce the proteotoxicity caused by these aberrant proteins. When proteasome activity is inadequate, ubiquitinated proteins are sorted into perinuclear aggresomes, which is a significant defense mechanism employed by cells to combat insufficient proteasome activity, hence mitigating the proteotoxic crisis. It has been demonstrated that phosphorylation of SQSTM1 is crucial in regulating misfolded protein aggregation and autophagic degradation. Although SQSTM1 S403 phosphorylation is essential for the autophagic degradation of ubiquitinated proteins, its significance in proteasome inhibition-induced aggresome formation is yet unknown. Herein, we investigated the influence of SQSTM1 S403 phosphorylation on the aggresome production of ubiquitinated proteins during proteasome suppression.
Methods: We examined the phosphorylation levels of SQSTM1 S403 or T269/S272 in cells after treated with proteasome inhibitors or/and autophagy inhibitors, by western blot and immunofluorescence. We detected the accumulation and aggresome formation of ubiquitinated misfolded proteins in cells treated with proteasome inhibition by western blot and immunofluorescence. Furthermore, we used SQSTM1 phosphorylation-associated kinase inhibitors and mutant constructs to confirm the regulation of different SQSTM1 phosphorylation in aggresome formation. We examined the cell viability using CCK-8 assay.
Results: Herein, we ascertained that phosphorylation of SQSTM1 S403 did not enhance the autophagic degradation of ubiquitinated proteins during proteasome inhibition. Proteasome inhibition suppresses the phosphorylation of SQSTM1 S403, which facilitated the aggresome production of polyubiquitinated proteins. Interestingly, we found proteasome inhibition-induced SQSTM1 T269/S272 phosphorylation inhibits the S403 phosphorylation. Suppressing S403 phosphorylation rescues the defective aggresome formation and protects cells from cell death caused by unphosphorylated SQSTM1 (T269/S272).
Conclusions: This study shows that inhibition of SQSTM1 S403 phosphorylation facilitates the aggresome formation of ubiquitinated proteins during proteasome dysfunction. SQSTM1 T269/S272 phosphorylation inhibits the S403 phosphorylation, boosting the aggresome formation of ubiquitinated protein and shielding cells from proteotoxic crisis.
Keywords SQSTM1 phosphorylation, Proteasome inhibition, Aggresome, Cell death, Aggrephagy
Address and Contact Information 1 Division of Abdominal Tumor Multimodality Treatment, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
2 Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
3 Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, Sichuan Province, China
* Corresponding author: zhangchenliang@wchscu.cn
Read full article at BMC

No.  86DOI: 10.1186/s11658-023-00497-y Volume 28 (2023) - 28:86
Title EFR3A: A NEW RAFT DOMAIN ORGANIZING PROTEIN?
Authors Magdalena Trybus1†, Anita Hryniewicz‐Jankowska1†, Karolina Wójtowicz2, Tomasz Trombik3, Aleksander Czogalla1* and Aleksander F. Sikorski4*
Abstract Background: Membrane rafts play a crucial role in the regulation of many important biological processes. Our previous data suggest that specific interactions of flotillins with MPP1 are responsible for membrane raft domain organization and regulation in erythroid cells. Interaction of the flotillin-based protein network with specific membrane components underlies the mechanism of raft domain formation and regulation, including in cells with low expression of MPP1.
Methods: We sought to identify other flotillin partners via the immobilized recombinant flotillin-2-based affinity approach and mass spectrometry technique. The results were further confirmed via immunoblotting and via co-immunoprecipitation. In order to study the effect of the candidate protein on the physicochemical properties of the plasma membrane, the gene was knocked down via siRNA, and fluorescence lifetime imaging microscopy and spot-variation fluorescence correlation spectroscopy was employed.
Results: EFR3A was identified as a candidate protein that interacts with flotillin-2. Moreover, this newly discovered interaction was demonstrated via overlay assay using recombinant EFR3A and flotillin-2. EFR3A is a stable component of the detergent-resistant membrane fraction of HeLa cells, and its presence was sensitive to the removal of cholesterol. While silencing the EFR3A gene, we observed decreased order of the plasma membrane of living cells or giant plasma membrane vesicles derived from knocked down cells and altered mobility of the raft probe, as indicated via fluorescence lifetime imaging microscopy and spot-variation fluorescence correlation spectroscopy. Moreover, silencing of EFR3A expression was found to disturb epidermal growth factor receptor and phospholipase C gamma phosphorylation and affect epidermal growth factor-dependent cytosolic Ca2+ concentration.
Conclusions: Altogether, our results suggest hitherto unreported flotillin-2-EFR3A interaction, which might be responsible for membrane raft organization and regulation. This implies participation of this interaction in the regulation of multiple cellular processes, including those connected with cell signaling which points to the possible role in human health, in particular human cancer biology.
Keywords Raft domain organization and regulation, Flotillins, Flotillin-2, EFR3A, Membrane order, FLIM, svFCS, EGFR
Address and Contact Information 1 Department of Cytobiochemistry, Faculty of Biotechnology, University of Wroclaw, F. Joliot‐Curie 14a, 50‐383 Wroclaw, Poland
2 Department of Biotransformation, Faculty of Biotechnology, University of Wroclaw, F. Joliot‐Curie 14a, 50‐383 Wroclaw, Poland
3 Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1, 20‐093 Lublin, Poland
4 Research and Development Center, Regional Specialist Hospital, Kamienskiego73a, 51‐154 Wroclaw, Poland
*Corresponding author: aleksander.czogalla@uwr.edu.pl; aleksander.sikorski@wssk.wroc.pl
Magdalena Trybus and Anita Hryniewicz-Jankowska equally contributed to this work.
Read full article at BMC

No.  87DOI: https://cmbl.biomedcentral.com/articles/10.1186/s11658-023-00498-x Volume 28 (2023) - 28:87
Title CADMIUM PROMOTES NONALCOHOLIC FATTY LIVER DISEASE BY INHIBITING INTERCELLULAR MITOCHONDRIAL TRANSFER
Authors Jian Sun1,2, Yan Chen1,2, Tao Wang1,2, Waseem Ali1,2, Yonggang Ma1,2, Yan Yuan1,2, Jianhong Gu1,2, Jianchun Bian1,2, Zongping Liu1,2 and Hui Zou1,2*
Abstract Mitochondrial transfer regulates intercellular communication, and mitochondria regulate cell metabolism and cell survival. However, the role and mechanism of mitochondrial transfer in Cd-induced nonalcoholic fatty liver disease (NAFLD) are unclear. The present study shows that mitochondria can be transferred between hepatocytes via microtubule-dependent tunneling nanotubes. After Cd treatment, mitochondria exhibit perinuclear aggregation in hepatocytes and blocked intercellular mitochondrial transfer. The different movement directions of mitochondria depend on their interaction with different motor proteins. The results show that Cd destroys the mitochondria-kinesin interaction, thus inhibiting mitochondrial transfer. Moreover, Cd increases the interaction of P62 with Dynactin1, promotes negative mitochondrial transport, and increases intracellular lipid accumulation. Mitochondria and hepatocyte co-culture significantly reduced Cd damage to hepatocytes and lipid accumulation. Thus, Cd blocks intercellular mitochondrial transfer by disrupting the microtubule system, inhibiting mitochondrial positive transport, and promoting their negative transport, thereby promoting the development of NAFLD.

Highlights:
1. Mitochondrial transfer between hepatocytes can be achieved through microtubule-dependent tunneling nanotubes
2. Cadmium inhibits the intercellular mitochondrial transfer
3. Inhibition of intercellular mitochondrial transfer mediates cadmium-induced nonalcoholic fatty liver disease.
4. Motor protein-dependent mitochondrial mobility is a target for cadmium to inhibit intercellular mitochondrial transfer
Keywords Cadmium, Intercellular mitochondrial transfer, Tunneling nanotubes, Nonalcoholic fatty liver disease
Address and Contact Information 1 College of Veterinary Medicine, Yangzhou University, Yangzhou, China
2 Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
*Corresponding author: zouhui@yzu.edu.cn
Read full article at BMC

No.  88DOI: 10.1186/s11658-023-00501-5 Volume 28 (2023) - 28:88
Title CELLULAR SENESCENCE IN SKELETAL DISEASE: MECHANISMS AND TREATMENT
Authors Xu He1†, Wei Hu1†, Yuanshu Zhang2†, Mimi Chen3, Yicheng Ding4, Huilin Yang1, Fan He1*, Qiaoli Gu1* and Qin Shi1,2*
Abstract The musculoskeletal system supports the movement of the entire body and provides blood production while acting as an endocrine organ. With aging, the balance of bone homeostasis is disrupted, leading to bone loss and degenerative diseases, such as osteoporosis, osteoarthritis, and intervertebral disc degeneration. Skeletal diseases have a profound impact on the motor and cognitive abilities of the elderly, thus creating a major challenge for both global health and the economy. Cellular senescence is caused by various genotoxic stressors and results in permanent cell cycle arrest, which is considered to be the underlying mechanism of aging. During aging, senescent cells (SnCs) tend to aggregate in the bone and trigger chronic inflammation by releasing senescence-associated secretory phenotypic factors. Multiple signalling pathways are involved in regulating cellular senescence in bone and bone marrow microenvironments. Targeted SnCs alleviate age-related degenerative diseases. However, the association between senescence and age-related diseases remains unclear. This review summarises the fundamental role of senescence in age-related skeletal diseases, highlights the signalling pathways that mediate senescence, and discusses potential therapeutic strategies for targeting SnCs.
Keywords Cellular senescence, Chronic infammation, Age-related orthopaedic diseases, Signalling pathways, Bone marrow
Address and Contact Information 1 Department of Orthopedics, The First Afliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu 215031, People’s Republic of China
2 Department of Orthopedics, Wuxi Ninth People’s Hospital Afliated to Soochow University, Wuxi, Jiangsu 214026, People’s Republic of China
3 Department of Orthopedics, Children Hospital of Soochow University, No. 92 Zhongnan Street, Suzhou, Jiangsu 215000, People’s Republic of China
4 Xuzhou Medical University, 209 Copper Mountain Road, Xuzhou 221004, People’s Republic of China *Correspondinga author: fanhe@suda.edu.cn; guqiaoli@suda.edu.cn; shiqin@suda.edu.cn
Xu He, Wei Hu and Yuanshu Zhang contributed equally to this work.
Read full article at BMC

No.  89DOI: 10.1186/s11658-023-00493-2 Volume 28 (2023) - 28:89
Title N6-METHYLADENOSINE-MODIFIED ONCOFETAL lncRNA MIR4435-2HG CONTRIBUTED TO STEMNESS FEATURES OF HEPATOCELLULAR CARCINOMA CELLS BY REGULATING rRNA 2′-O METHYLATION
Authors Yiqing Zhu1†, Bang Xiao1†, Meng Liu1†, Meiting Chen1†, Ningqi Xia1, Haiyan Guo2, Jinfeng Huang1*, Zhiyong Liu3* and Fang Wang1*
Abstract Background: The unique expression pattern endows oncofetal genes with great value in cancer diagnosis and treatment. However, only a few oncofetal genes are available for clinical use and the underlying mechanisms that drives the fetal-like reprogramming of cancer cells remain largely unknown.
Methods: Microarray assays and bioinformatic analyses were employed to screen for potential oncofetal long non-coding RNAs (lncRNAs) in hepatocellular carcinoma (HCC). The expression levels of MIR4435-2HG, NOP58 ribonucleoprotein (NOP58), insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) and stem markers were detected by quantitative polymerase chain reaction. The 2′-O-methylation (2′-O-Me) status of rRNA were detected through reverse transcription at low dNTP concentrations followed by PCR. The regulation of MIR4435-2HG by IGF2BP1 was explored by RNA immunoprecipitation (RIP), methylated RIP (MeRIP) and dual-luciferase assays. The interaction between MIR4435-2HG and NOP58 was investigated by RNA Pulldown, RIP and protein stability assays. In vitro and in vivo function assays were performed to detect the roles of MIR4435-2HG/NOP58 in HCC.
Results: MIR4435-2HG was an oncofetal lncRNA associated with poor prognosis in HCC. Functional experiments showed that overexpression of MIR4435-2HG remarkably enhanced the stem-cell properties of HCC cells, promoting tumorigenesis in vitro and in vivo. Mechanically, MIR4435-2HG directly bound NOP58 and IGF2BP1. IGF2BP1 upregulated MIR4435-2HG expression in HCC through N6-methyladenosine (m6A) modification. Moreover, MIR4435-2HG protected NOP58 from degradation, which raised rRNA 2’-O-Me levels and promoted internal ribosome entry site (IRES)-dependent translation of oncogenes.
Conclusions: This study identified an oncofetal lncRNA MIR4435-2HG, characterized the role of MIR4435-2HG/NOP58 in stemness maintenance and proliferation of HCC cells, and confirmed m6A as a ‘driver’ that reactivated MR4435-2HG expression in HCC.
Keywords Oncofetal lncRNA, Hepatocellular carcinoma, Stemness maintenance, rRNA 2′-O-methylation, N6-methyladenosine
Address and Contact Information 1 Department of Medical Genetics, Naval Medical University, Shanghai 200433, China
2 Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Center for Specialty Strategy Research of Shanghai Jiao Tong University China Hospital Development Institute, Shanghai 200011, China
3 Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
*Corresponding author: jfhuang@smmu.edu.cn; chyang@sibs.ac.cn; wfsjz@smmu.edu.cn
Yiqing Zhu, Bang Xiao, Meng Liu, Meiting Chen contributed equally.
Read full article at BMC

No.  90DOI: 10.1186/s11658-023-00506-0 Volume 28 (2023) - 28:90
Title GCN5L1 REGULATES PULMONARY SURFACTANT PRODUCTION BY MODULATING LAMELLAR BODY BIOGENESIS AND TRAFFICKING IN MOUSE ALVEOLAR EPITHELIAL CELLS
Authors Wenqin Xu1,2,3†, Xiaocui Ma4†, Qing Wang1,2,3†, Jingjing Ye1,2,3†, Nengqian Wang5, Zhenzhen Ye5and Tianbing Chen1,2,3*
Abstract Background: The pulmonary surfactant that lines the air–liquid surface within alveoli is a protein–lipid mixture essential for gas exchange. Surfactant lipids and proteins are synthesized and stored in the lamellar body (LB) before being secreted from alveolar type II (AT2) cells. The molecular and cellular mechanisms that regulate these processes are incompletely understood. We previously identified an essential role of general control of amino acid synthesis 5 like 1 (GCN5L1) and the biogenesis of lysosome-related organelle complex 1 subunit 1 (BLOS1) in surfactant system development in zebrafish. Here, we explored the role of GCN5L1 in pulmonary surfactant regulation.
Method: GCN5L1 knockout cell lines were generated with the CRISPR/Cas9 system. Cell viability was analyzed by MTT assay. Released surfactant proteins were measured by ELISA. Released surfactant lipids were measured based on coupled enzymatic reactions. Gene overexpression was mediated through lentivirus. The RNA levels were detected through RNA-sequencing (RNA-seq) and quantitative reverse transcription (qRT)- polymerase chain reaction (PCR). The protein levels were detected through western blotting. The cellular localization was analyzed by immunofluorescence. Morphology of the lamellar body was analyzed through transmission electron microscopy (TEM), Lysotracker staining, and BODIPY phosphatidylcholine labeling.
Results: Knocking out GCN5L1 in MLE-12 significantly decreased the release of surfactant proteins and lipids. We detected the downregulation of some surfactant-related genes and misregulation of the ROS–Erk–Foxo1–Cebpα axis in mutant cells. Modulating the activity of the axis or reconstructing the mitochondrial expression of GCN5L1 could partially restore the expression of these surfactant-related genes. We further showed that MLE-12 cells contained many LB-like organelles that were lipid enriched and positive for multiple LB markers. These organelles were smaller in size and accumulated in the absence of GCN5L1, indicating both biogenesis and trafficking defects. Accumulated endogenous surfactant protein (SP)-B or exogenously expressed SP-B/SP-C in adenosine triphosphate-binding cassette transporterA3 (ABCA3)-positive organelles was detected in mutant cells. GCN5L1 localized to the mitochondria and LBs. Reconstruction of mitochondrial GCN5L1 expression rescued the organelle morphology but failed to restore the trafficking defect and surfactant release, indicating specific roles associated with different subcellular localizations.
Conclusions: In summary, our study identified GCN5L1 as a new regulator of pulmonary surfactant that plays a role in the biogenesis and positioning/trafficking of surfactant-containing LBs.
Keywords Pulmonary surfactant, Type II alveolar epithelial cell, Lamellar body, Trafficking, GCN5L1
Address and Contact Information 1 Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China
2 Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
3 Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, China
4 Henan Clinical Research Center of Childhood Diseases, Children’s Hospital Afliated to Zhengzhou University, Zhengzhou, China
5 Department of Pediatrics, Yijishan Hospital of Wannan Medical College, Wuhu, China
*Corresponding author: ctb0410021@126.com
Wenqin Xu, Xiaocui Ma, Qing Wang and Jingjing Ye contribute equally to the article.
Read full article at BMC

No.  91DOI: 10.1186/s11658-023-00495-0 Volume 28 (2023) - 28:91
Title Irf7 REGULATES THE EXPRESSION OF Srg3 AND FERROPTOSIS AXIS AGGRAVATED SEPSIS-INDUCED ACUTE LUNG INJURY
Authors Xinyu Ling1†, Shiyou Wei2†, Dandan Ling3, Siqi Cao4, Rui Chang5, Qiuyun Wang6* and Zhize Yuan1*
Abstract Objective: To investigate the mechanism of action of Srg3 in acute lung injury caused by sepsis.
Methods: First, a sepsis-induced acute lung injury rat model was established using cecal ligation and puncture (CLP). RNA sequencing (RNA-seq) was used to screen for highly expressed genes in sepsis-induced acute lung injury (ALI), and the results showed that Srg3 was significantly upregulated. Then, SWI3-related gene 3 (Srg3) was knocked down using AAV9 vector in vivo, and changes in ALI symptoms in rats were analyzed. In vitro experiments were conducted by establishing a cell model using lipopolysaccharide (LPS)-induced BEAS-2B cells and coculturing them with phorbol 12-myristate 13-acetate (PMA)-treated THP-1 cells to analyze macrophage polarization. Next, downstream signaling pathways regulated by Srg3 and transcription factors involved in regulating Srg3 expression were analyzed using the KEGG database. Finally, gain-of-loss functional validation experiments were performed to analyze the role of downstream signaling pathways regulated by Srg3 and transcription factors involved in regulating Srg3 expression in sepsis-induced acute lung injury.
Results: Srg3 was significantly upregulated in sepsis-induced acute lung injury, and knocking down Srg3 significantly improved the symptoms of ALI in rats. Furthermore, in vitro experiments showed that knocking down Srg3 significantly weakened the inhibitory effect of LPS on the viability of BEAS-2B cells and promoted alternative activation phenotype (M2) macrophage polarization. Subsequent experiments showed that Srg3 can regulate the activation of the NF-κB signaling pathway and promote ferroptosis. Specific activation of the NF-κB signaling pathway or ferroptosis significantly weakened the effect of Srg3 knockdown. It was then found that Srg3 can be transcriptionally activated by interferon regulatory factor 7 (Irf7), and specific inhibition of Irf7 significantly improved the symptoms of ALI.
Conclusions: Irf7 transcriptionally activates the expression of Srg3, which can promote ferroptosis and activate classical activation phenotype (M1) macrophage polarization by regulating the NF-κB signaling pathway, thereby exacerbating the symptoms of septic lung injury.
Keywords Sepsis-induced acute lung injury, Srg3, Irf7, Ferroptosis, NF-κB signaling pathway
Address and Contact Information 1 Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
2 Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
3 Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
4 School of Clinical Medicine, Weifang Medical University, Weifang 261053, Shandong, China
5 Medical Department, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
6 Department of Anesthesiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
*Corresponding author: drwangqiuyun@126.com; yuanzhize0402@tongji.edu.cn
Xinyu Ling and Shiyou Wei are equally contributed to this study.
Read full article at BMC

No.  92DOI: 10.1186/s11658-023-00505-1 Volume 28 (2023) - 28:92
Title EXOSOMAL miR-155-5p DRIVES WIDESPREAD MACROPHAGE M1 POLARIZATION IN HYPERVIRULENT Klebsiella pneumoniae-INDUCED ACUTE LUNG INJURY VIA THE MSK1/p38-MAPK AXIS
Authors Yihan Xu1, Chunying Zhang1, Danni Cai2, Rongping Zhu1 and Yingping Cao1*
Abstract Background: Hypervirulent Klebsiella pneumoniae (hvKp) infection-induced sepsis-associated acute lung injury (ALI) has emerged as a significant clinical challenge. Increasing evidence suggests that activated inflammatory macrophages contribute to tissue damage in sepsis. However, the underlying causes of widespread macrophage activation remain unclear.
Methods: BALB/c mice were intravenously injected with inactivated hvKp (iHvKp) to observe lung tissue damage, inflammation, and M1 macrophage polarization. In vitro, activated RAW264.7 macrophage-derived exosomes (iHvKp-exo) were isolated and their role in ALI formation was investigated. RT-PCR was conducted to identify changes in exosomal miRNA. Bioinformatics analysis and dual-luciferase reporter assays were performed to validate MSK1 as a direct target of miR-155-5p. Further in vivo and in vitro experiments were conducted to explore the specific mechanisms involved.
Results: iHvKp successfully induced ALI in vivo and upregulated the expression of miR-155-5p. In vivo, injection of iHvKp-exo induced inflammatory tissue damage and macrophage M1 polarization. In vitro, iHvKp-exo was found to promote macrophage inflammatory response and M1 polarization through the activation of the p38-MAPK pathway. RT-PCR revealed exposure time-dependent increased levels of miR-155-5p in iHvKp-exo. Dual-luciferase reporter assays confirmed the functional role of miR-155-5p in mediating iHvKp-exo effects by targeting MSK1. Additionally, inhibition of miR-155-5p reduced M1 polarization of lung macrophages in vivo, resulting in decreased lung injury and inflammation induced by iHvKp-exo or iHvKp.
Conclusions: The aforementioned results indicate that exosomal miR-155-5p drives widespread macrophage inflammation and M1 polarization in hvKp-induced ALI through the MSK1/p38-MAPK Axis.
Keywords Hypervirulent Klebsiella pneumoniae, Acute lung injury, Macrophage, Exosome, miR-155-5p
Address and Contact Information 1 Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou 350001, People’s Republic of China
2 Central Laboratory, Fujian Medical University Union Hospital, Fuzhou 350001, People’s Republic of China
*Corresponding author: caoyingping@aliyun.com
Read full article at BMC

No.  93DOI: 10.1186/s11658-023-00510-4 Volume 28 (2023) - 28:93
Title DISRUPTED CARDIAC FIBROBLAST BCAA CATABOLISM CONTRIBUTES TO DIABETIC CARDIOMYOPATHY VIA A PERIOSTIN/NAP1L2/SIRT3 AXIS
Authors Qing‐Bo Lu1,2†, Xiao Fu1†, Yao Liu3†, Zi‐Chao Wang4, Shi‐Yi Liu1, Yu‐Chao Li1 and Hai‐Jian Sun1,4*
Abstract Background: Periostin is an extracellular matrix protein that plays a critical role in cell fate determination and tissue remodeling, but the underlying role and mechanism of periostin in diabetic cardiomyopathy (DCM) are far from clear. Thus, we aimed to clarify the mechanistic participation of periostin in DCM.
Methods: The expression of periostin was examined in DCM patients, diabetic mice and high glucose (HG)-exposed cardiac fibroblasts (CF). Gain- and loss-of-function experiments assessed the potential role of periostin in DCM pathogenesis. RNA sequencing was used to investigate the underlying mechanisms of periostin in DCM.
Results: A mouse cytokine antibody array showed that the protein expression of periostin was most significantly upregulated in diabetic mouse heart, and this increase was also observed in patients with DCM or HG-incubated CF. Periostin-deficient mice were protected from diabetes-induced cardiac dysfunction and myocardial damage, while overexpression of periostin held the opposite effects. Hyperglycemia stimulated the expression of periostin in a TGF-β/Smad-dependent manner. RNA sequencing results showed that periostin upregulated the expression of nucleosome assembly protein 1-like 2 (NAP1L2) which recruited SIRT3 to deacetylate H3K27ac on the promoters of the branched-chain amino acid (BCAA) catabolism-related enzymes BCAT2 and PP2Cm, resulting in BCAA catabolism impairment. Additionally, CF-derived periostin induced hypertrophy, oxidative injury and inflammation in primary cardiomyocytes. Finally, we identified that glucosyringic acid (GA) specifically targeted and inhibited periostin to ameliorate DCM.
Conclusion: Overall, manipulating periostin expression may function as a promising strategy in the treatment of DCM.
Keywords Periostin, Diabetes, Cardiomyopathy, BCAA catabolism, Glucosyringic acid
Address and Contact Information 1 Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
2 Department of Endocrine, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi 214125, China
3 Department of Cardiac Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing 210000, Jiangsu, China
4 State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
*Corresponding author: haijsunjiangnan@jiangnan.edu.cn
Qing-Bo Lu, Xiao Fu and Yao Liu contributed equally.
Read full article at BMC

No.  94DOI: 10.1186/s11658-023-00512-2 Volume 28 (2023) - 28:94
Title Correction: Patterns of human and porcine gammaherpesvirus-encoded BILF1 receptor endocytosis
Authors Maša Mavri1, Sanja Glišić2, Milan Senćanski2, Milka Vrecl1, Mette M. Rosenkilde3, Katja Spiess3,4 and Valentina Kubale1*
Abstract Correction : Cellular & Molecular Biology Letters (2023) 28:14 https://doi.org/10.1186/s11658-023-00427-y

Following publication of the original article [1], the authors updated the Funding section.

The original article has been corrected.

Published: 24 November 2023

Reference
1. Mavri M, Glišić S, Senćanski M, Vrecl M, Rosenkilde MM, Spiess K, Kubale V. Patterns of human and porcine gammaherpesvirus-encoded BILF1 receptor endocytosis. Cell Mol Biol Lett. 2023;28:14. https://doi.org/10.1186/ s11658-023-00427-y.

Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional afliations.
Keywords
Address and Contact Information 1 Institute for Preclinical Sciences, Veterinary Faculty, Ljubljana, Slovenia
2 Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
3 Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
4 Present Address: Department of Virus and Microbiological Special Diagnostics, Statens Serum Institute, Copenhagen, Denmark
*Corresponding author: valentina.kubale@vf.uni-lj.si
The original article can be found online at https://doi.org/10.1186/s11658-023-00427-y.
Read full article at BMC

No.  95DOI: 10.1186/s11658-023-00513-1 Volume 28 (2023) - 28:95
Title ASTAXANTHIN ALLEVIATES PM2.5-INDUCED CARDIOMYOCYTE INJURY VIA INHIBITING FERROPTOSIS
Authors Jingyi Ren1†, Bowen Yin1†, Zihao Guo2†, Xiaoya Sun1, Huanting Pei1, Rui Wen1, Ziyi Wang2, Siqi Zhu1, Jinshi Zuo1, Yadong Zhang1 and Yuxia Ma1*
Abstract Background: Long-term exposure of humans to air pollution is associated with an increasing risk of cardiovascular diseases (CVDs). Astaxanthin (AST), a naturally occurring red carotenoid pigment, was proved to have multiple health benefits. However, whether or not AST also exerts a protective effect on fine particulate matter (PM2.5)-induced cardiomyocyte damage and its underlying mechanisms remain unclear.
Methods: In vitro experiments, the H9C2 cells were subjected to pretreatment with varying concentrations of AST, and then cardiomyocyte injury model induced by PM2.5 was established. The cell viability and the ferroptosis-related proteins expression were measured in different groups. In vivo experiments, the rats were pretreated with different concentrations of AST for 21 days. Subsequently, a rat model of myocardial PM2.5 injury was established by intratracheal instillation every other day for 1 week. The effects of AST on myocardial tissue injury caused by PM2.5 indicating by histological, serum, and protein analyses were examined.
Results: AST significantly ameliorated PM2.5-induced myocardial tissue injury, inflammatory cell infiltration, the release of inflammatory factors, and cardiomyocyte H9C2 cell damage. Mechanistically, AST pretreatment increased the expression of SLC7A11, GPX4 and down-regulated the expression of TfR1, FTL and FTH1 in vitro and in vivo.
Conclusions: Our study suggest that ferroptosis plays a significant role in the pathogenesis of cardiomyocyte injury induced by PM2.5. AST may serve as a potential therapeutic agent for mitigating cardiomyocyte injury caused by PM2.5 through the inhibition of ferroptosis.
Keywords Astaxanthin, Cardiomyocyte injury, Cardiovascular diseases, PM2.5, Ferroptosis
Address and Contact Information 1 Department of Nutrition and Food Hygiene, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang 050017, China
2 Undergraduate of College of Public Health, Hebei Medical University, Shijiazhuang 050017, China
*Corresponding author: mayuxia@hebmu.edu.cn
Jingyi Ren, Bowen Yin and Zihao Guo have contributed equally to this work.
Read full article at BMC

No.  96DOI: 10.1186/s11658-023-00508-y Volume 28 (2023) - 28:96
Title SYNTHETIC CRISPR/dCas9-KRAB SYSTEM DRIVEN BY SPECIFIC PSA PROMOTER SUPPRESSES MALIGNANT BIOLOGICAL BEHAVIOR OF PROSTATE CANCER CELLS THROUGH NEGATIVE FEEDBACK INHIBITION OF PSA EXPRESSION
Authors Yi Yang1, Hongbing Mei1, Xiaohong Han1, Xintao Zhang1, Jianli Cheng1, Zhongfu Zhang1, Han Wang1 and Haixia Xu2*
Abstract PSA is a type of proto-oncogene that is specifically and highly expressed in embryonic and prostate cancer cells, but not expressed in normal prostate tissue cells. The specific expression of prostate-specific antigen (PSA) is found to be related with the conditional transcriptional regulation of its promoter. Clustered regularly interspaced short palindromic repeats (CRISPR)-dCas9-KRAB is a newly developed transcriptional regulatory system that inhibits gene expression by interupting the DNA transcription process. Induction of CRISPR-dCas9-KRAB expression through the PSA promoter may help feedback inhibition of cellular PSA gene expression via single guide RNA (sgRNA), thereby monitoring and suppressing the malignant state of tumor cells. In this study, we examined the transcriptional activity of the PSA promoter in different prostate cancer cells and normal prostate epithelial cells and determined that it is indeed a prostate cancer cell-specific promoter.Then we constructed the CRISPR-dCas9-KRAB system driven by the PSA promoter, which can inhibit PSA gene expression in the prostate cancer cells at the transcriptional level, and therefore supress the malignant growth and migration of prostate cancer cells and promote their apoptosis in vitro. This study provides a potentially effective anti-cancer strategy for gene therapy of prostate cancer.
Keywords CRISPR-dCas9-KRAB, Prostate cancer, PSA, Gene therapy
Address and Contact Information 1 Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
2 Department of Medical Oncology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
*Corresponding author: 182199558@qq.com
Read full article at BMC

No.  97DOI: 10.1186/s11658-023-00511-3 Volume 28 (2023) - 28:97
Title KNOCK-IN MICE EXPRESSING A HUMANIZED ARACHIDONIC ACID 15-LIPOXYGENASE (Alox15) CARRY A PARTLY DYSFUNCTIONAL ERYTHROPOIETIC SYSTEM
Authors Florian Reisch1,2,3, Dagmar Heydeck1, Marjann Schäfer1,2, Michael Rothe3, Jiaxing Yang1, Sabine Stehling1, Gerhard P. Püschel2 and Hartmut Kuhn1*
Abstract Arachidonic acid 15-lipoxygenases (ALOX15) play a role in mammalian erythropoiesis but they have also been implicated in inflammatory processes. Seven intact Alox genes have been detected in the mouse reference genome and the mouse Alox15 gene is structurally similar to the orthologous genes of other mammals. However, mouse and human ALOX15 orthologs have different functional characteristics. Human ALOX15 converts C20 polyenoic fatty acids like arachidonic acid mainly to the n-6 hydroperoxide. In contrast, the n-9 hydroperoxide is the major oxygenation product formed by mouse Alox15. Previous experiments indicated that Leu353Phe exchange in recombinant mouse Alox15 humanized the catalytic properties of the enzyme. To investigate whether this functional humanization might also work in vivo and to characterize the functional consequences of mouse Alox15 humanization we generated Alox15 knock-in mice (Alox15-KI), in which the Alox15 gene was modified in such a way that the animals express the arachidonic acid 15-lipoxygenating Leu353Phe mutant instead of the arachidonic acid 12-lipoxygenating wildtype enzyme. These mice develop normally, they are fully fertile but display modified plasma oxylipidomes. In young individuals, the basic hematological parameters were not different when Alox15-KI mice and outbred wildtype controls were compared. However, when growing older male Alox15-KI mice develop signs of dysfunctional erythropoiesis such as reduced hematocrit, lower erythrocyte counts and attenuated hemoglobin concentration. These differences were paralleled by an improved ex vivo osmotic resistance of the peripheral red blood cells. Interestingly, such differences were not observed in female individuals suggesting gender specific effects. In summary, these data indicated that functional humanization of mouse Alox15 induces defective erythropoiesis in aged male individuals.
Keywords Eicosanoids, Lipid peroxidation, Oxidative stress, Polyenoic fatty acids, Erythropoiesis
Address and Contact Information 1 Department of Biochemistry, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
2 Institute for Nutritional Sciences, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
3 Lipidomix GmbH, Robert-Rössle-Straße 10, 13125 Berlin, Germany
*Corresponding author: hartmut.kuehn@charite.de
Read full article at BMC

No.  98DOI: 10.1186/s11658-023-00504-2 Volume 28 (2023) - 28:98
Title POTENTIAL USE OF iPSCs FOR DISEASE MODELING, DRUG SCREENING, AND CELL-BASED THERAPY FOR ALZHEIMER’S DISEASE
Authors Hany E. Marei1*, Muhammad Umar Aslam Khan2,3 and Anwarul Hasan3
Abstract Alzheimer’s disease (AD) is a chronic illness marked by increasing cognitive decline and nervous system deterioration. At this time, there is no known medication that will stop the course of Alzheimer’s disease; instead, most symptoms are treated. Clinical trial failure rates for new drugs remain high, highlighting the urgent need for improved AD modeling for improving understanding of the underlying pathophysiology of disease and improving drug development. The development of induced pluripotent stem cells (iPSCs) has made it possible to model neurological diseases like AD, giving access to an infinite number of patient-derived cells capable of differentiating neuronal fates. This advance will accelerate Alzheimer’s disease research and provide an opportunity to create more accurate patient-specific models of Alzheimer’s disease to support pathophysiological research, drug development, and the potential application of stem cell-based therapeutics. This review article provides a complete summary of research done to date on the potential use of iPSCs from AD patients for disease modeling, drug discovery, and cell-based therapeutics. Current technological developments in AD research including 3D modeling, genome editing, gene therapy for AD, and research on familial (FAD) and sporadic (SAD) forms of the disease are discussed. Finally, we outline the issues that need to be elucidated and future directions for iPSC modeling in AD.
Keywords Alzheimer’s diseases, Induced pluripotent stem cells, iPSCs, Disease modeling, Drug development, Mechanism of diseases, Regenerative medicine, Cell-based therapies
Address and Contact Information 1 Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt
2 Biomedical Research Center, Qatar University, 2713 Doha, Qatar
3 Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, Qatar
*Corresponding author: hanymarei@mans.edu.eg
Read full article at BMC

No.  99DOI: 10.1186/s11658-023-00507-z Volume 28 (2023) - 28:99
Title GLOBAL RESEARCH TRENDS IN CRISPR-RELATED TECHNOLOGIES ASSOCIATED WITH EXTRACELLULAR VESICLES FROM 2015 TO 2022: A BIBLIOMETRIC, DYNAMIC, AND VISUALIZED STUDY
Authors Jianjing Lin1†, Shicheng Jia1,2†, Zilu Jiao1†, Jiayou Chen1,2, Wei Li1*, Fuyang Cao3* and Xintao Zhang1*
Abstract Purpose: This study aims to explore the emerging trends, dynamic development, and research hotspots of clustered regularly interspaced short palindromic repeats (CRISPR) technology associated with extracellular vesicles during the past 7 years and demonstrate them by visualization.
Methods: A total of 219 records related to CRISPR technology associated with extracellular vesicles from 2015 to 2022 in the Web of Science Core Collection (WoSCC) database were collected. R language, VOSviewer, CiteSpace, and GraphpadPrism software packages were used to analyze the history of this research, the general characteristics of the literature, and keywords. Finally, the hotspots and latest trends in CRISPR technology associated with extracellular vesicles are predicted.
Results: A total of 219 articles were collected for this study. The production of publications about CRISPR technology associated with extracellular vesicles has increased annually. Researchers from China, the USA, and Germany made the most important contributions to this trend, while RLUK Research Libraries UK offers the largest amount of literature in this field. Shenzhen University, Nanjing Medicine University, and Peking University exhibited the closest cooperation. Additionally, active topics burst during different periods, as identified according to 317 keywords belonging to 39 disciplines. Keywords were clustered into seven research subareas, namely exosome, nanovesicles, DNA, gene editing, gene therapy, cancer therapy, and endometrial stromal cells. The alluvial map of keywords reveals that the most enduring concepts are gene therapy, nanovesicles, etc., while the emerging keywords are genome, protein delivery, plasma, etc.
Conclusions: We reviewed 219 previous publications and conducted the first bibliometric study of CRISPR technology related to extracellular vesicles from 2015 to 2022. This comprehensive summary constructed a knowledge map and demonstrates the trends in this area. The current trends and potential hotpots for this topic are also identified, which will be a great help for researchers in the future.
Keywords CRISPR-related technology, Extracellular vesicles, Bibliometric, Visualization
Address and Contact Information 1 Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
2 Shantou University Medical College, Shantou, Guangdong, China
3 Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
*Corresponding author: lwjnu2003@126.com; drcao0603@163.com; zhangxintao@sina.com
Jianjing Lin, Shicheng Jia, and Zilu Jiao contribute equally to this work and share first authorship.
Read full article at BMC

No.  100DOI: 10.1186/s11658-023-00503-3 Volume 28 (2023) - 28:100
Title METABOLITE ITACONATE IN HOST IMMUNOREGULATION AND DEFENSE
Authors Wenchang Yang1,3†, Yaxin Wang2†, Kaixiong Tao1 and Ruidong Li1*
Abstract Metabolic states greatly influence functioning and differentiation of immune cells. Regulating the metabolism of immune cells can effectively modulate the host immune response. Itaconate, an intermediate metabolite derived from the tricarboxylic acid (TCA) cycle of immune cells, is produced through the decarboxylation of cis-aconitate by cis-aconitate decarboxylase in the mitochondria. The gene encoding cis-aconitate decarboxylase is known as immune response gene 1 (IRG1). In response to external proinflammatory stimulation, macrophages exhibit high IRG1 expression. IRG1/itaconate inhibits succinate dehydrogenase activity, thus influencing the metabolic status of macrophages. Therefore, itaconate serves as a link between macrophage metabolism, oxidative stress, and immune response, ultimately regulating macrophage function. Studies have demonstrated that itaconate acts on various signaling pathways, including Keap1-nuclear factor E2-related factor 2-ARE pathways, ATF3–IκBζ axis, and the stimulator of interferon genes (STING) pathway to exert antiinflammatory and antioxidant effects. Furthermore, several studies have reported that itaconate affects cancer occurrence and development through diverse signaling pathways. In this paper, we provide a comprehensive review of the role IRG1/itaconate and its derivatives in the regulation of macrophage metabolism and functions. By furthering our understanding of itaconate, we intend to shed light on its potential for treating inflammatory diseases and offer new insights in this field.
Keywords Itaconate, Itaconate derivative, Immunometabolism, Defense
Address and Contact Information 1 Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan 430022, Hubei, China
2 Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
3 Department of Gastrointestinal Surgery, Afliated Hospital of Qingdao University, Qingdao, China
*Corresponding author: liruidong@hust.edu.cn
Wenchang Yang and Yaxin Wang contributed equally to this paper.
Read full article at BMC

No.  101DOI: 10.1186/s11658-023-00515-z Volume 28 (2023) - 28:101
Title METTL3/YTHDC1-MEDICATED m6A MODIFICATION OF circRNA3634 REGULATES THE PROLIFERATION AND DIFFERENTIATION OF ANTLER CHONDROCYTES BY miR-124486-5-MAPK1 AXIS
Authors Mengmeng Song1, Haibo Yao1, Zitong Sun1, Danyang Chen1, Xiwen Xu1, Guohui Long1*, Lei Wu1* and Wei Hu1*
Abstract Background: The deer antler, a remarkable mammalian appendage, has a growth rate surpassing that of any other known osseous organ. Emerging evidence indicates that circRNA and MAPK1 play critical roles in chondrocytes. Thus, exploration of their functions in antler chondrocytes will help us to understand the mechanism regulating the rapid antler growth.
Methods: qRT-PCR, western blot, and immunohistochemistry were used to assess the expression of mRNAs and proteins. CCK-8, EdU, Cell migration, ALP activity detection, and ALP staining examined the effects of MAPK1 in antler chondrocytes. FISH, RIP, and luciferase assays were performed to evaluate the interactions among circRNA3634/MAPK1 and miR-124486-5. RIP and RAP assays proved the binding interaction between circRNA3634 and RBPs. Me-RIP was used to determine the m6A methylation modification of circRNA3634.
Results: This study revealed high MAPK1 expression in antler cartilage tissue. Overexpression of MAPK1 promoted the proliferation, migration, and differentiation of antler chondrocytes and increased the expression of MAPK3, RAF1, MEK1, RUNX2, and SOX9. The silencing of MAPK1 had the opposite effect. CircRNA3634 was found to act as a molecular sponge for miR-124486-5, leading to increased MAPK1 expression and enhanced proliferation and migration of antler chondrocytes through competitive miR-124486-5 binding. We discovered that METTL3 mediates m6A modification near the splicing site of circRNA3634 and is involved in the proliferation and differentiation of antler chondrocytes. The m6A reader YTHDC1 facilitated the nuclear export of circRNA3634 in an m6A-dependent manner. Our results indicate that m6A-modified circRNA3634 promotes the proliferation of antler chondrocytes by targeting MAPK1 and show that the nuclear export of circRNA3634 is related to the expression of YTHDC1, suggesting that circRNA3634 could represent a critical regeneration marker for the antler.
Conclusions: Our results revealed a novel m6A-modified circRNA3634 promoted the proliferation and differentiation of antler chondrocytes by regulating MAPK1. The nuclear export of circRNA3634 was related to the expression of YTHDC1.
Keywords Antler chondrocyte, Cell proliferation, MAPK1, circRNA3634, m6A, YTHDC1
Address and Contact Information 1 College of Life Science, Jilin Agriculture University, Changchun 130118, China
*Corresponding author: ghlong@jlau.edu.cn; 837660996@qq.com; huweilab@126.com
Read full article at BMC

No.  102DOI: 10.1186/s11658-023-00488-z Volume 28 (2023) - 28:102
Title REGULATOR OF G PROTEIN SIGNALING PROTEIN 6 ALLEVIATES ACUTE LUNG INJURY BY INHIBITING INFLAMMATION AND PROMOTING CELL SELF-RENEWAL IN MICE
Authors Juan Song1,2,3,4, Miao Li2,4, Cuicui Chen1,2,4, Jian Zhou1,2,4, Linlin Wang2,4, Yu Yan2,4, Jun She2,4*, Lin Tong2,3,4* and Yuanlin Song1,2,3,4*
Abstract Background: Acute respiratory distress syndrome (ARDS) is a disease with high mortality and morbidity. Regulator of G protein signaling protein 6 (RGS6), identified as a tumor suppressor gene, has received increasing attention owing to its close relationship with oxidative stress and inflammation. However, the association between ARDS and RGS6 has not been reported.
Methods: Congruously regulated G protein-coupled receptor (GPCR)-related genes and differentially expressed genes (DEGs) in an acute lung injury (ALI) model were identified, and functional enrichment analysis was conducted. In an in vivo study, the effects of RGS6 knockout were studied in a mouse model of ALI induced by lipopolysaccharide (LPS). HE staining, ELISA, and immunohistochemistry were used to evaluate pathological changes and the degree of inflammation. In vitro, qRT‒PCR, immunofluorescence staining, and western blotting were used to determine the dynamic changes in RGS6 expression in cells. The RGS6 overexpression plasmid was constructed for transfection. qRT‒PCR was used to assess proinflammatory factors transcription. Western blotting and flow cytometry were used to evaluate apoptosis and reactive oxygen species (ROS) production. Organoid culture was used to assess the stemness and self-renewal capacity of alveolar epithelial type II cells (AEC2s).
Results: A total of 110 congruously regulated genes (61 congruously upregulated and 49 congruously downregulated genes) were identified among GPCR-related genes and DEGs in the ALI model. RGS6 was downregulated in vivo and in vitro in the ALI model. RGS6 was expressed in the cytoplasm and accumulated in the nucleus after LPS stimulation. Compared with the control group, we found higher mortality, more pronounced body weight changes, more serious pulmonary edema and pathological damage, and more neutrophil infiltration in the RGS6 knockout group upon LPS stimulation in vivo. Moreover, AEC2s loss was significantly increased upon RGS6 knockout. Organoid culture assays showed slower alveolar organoid formation, fewer alveolar organoids, and impaired development of new structures after passaging upon RGS6 knockout. In addition, RGS6 overexpression decreased ROS production as well as proinflammatory factor transcription in macrophages and decreased apoptosis in epithelial cells.
Conclusions: RGS6 plays a protective role in ALI not only in early inflammatory responses but also in endogenous lung stem cell regeneration.
Keywords Acute lung injury, Regulator of G protein signaling 6, Infammation, Cell- renewal, Apoptosis
Address and Contact Information 1 Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
2 Shanghai Key Laboratory of Lung Infammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
3 Department of Pulmonary Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian 361000, China
4 Shanghai Respiratory Research Institute, Shanghai 200032, China
*Corresponding author: she.jun@zs-hospital.sh.cn; tong.lin@zs-hospital.sh.cn; song.yuanlin@zs-hospital.sh.cn
Read full article at BMC

No.  103DOI: 10.1186/s11658-023-00514-0 Volume 28 (2023) - 28:103
Title ROLE OF N6-METHYLADENOSINE METHYLATION IN GLIOMA: RECENT INSIGHTS AND FUTURE DIRECTIONS
Authors Chunlin Li1†, Bowen Li2†, Hui Wang3, Linglong Qu2, Hui Liu4, Chao Weng5,6, Jinming Han7* and Yuan Li8,9*
Abstract Glioma is the most pervasive intracranial tumor in the central nervous system (CNS), with glioblastoma (GBM) being the most malignant type having a highly heterogeneous cancer cell population. There is a significantly high mortality rate in GBM patients. Molecular biomarkers related to GBM malignancy may have prognostic values in predicting survival outcomes and therapeutic responses, especially in patients with high-grade gliomas. In particular, N6-methyladenine (m6A) mRNA modification is the most abundant form of post-transcriptional RNA modification in mammals and is involved in regulating mRNA translation and degradation. Cumulative findings indicate that m6A methylation plays a crucial part in neurogenesis and glioma pathogenesis. In this review, we summarize recent advances regarding the functional significance of m6A modification and its regulatory factors in glioma occurrence and progression. Significant advancement of m6A methylation-associated regulators as potential therapeutic targets is also discussed.
Keywords N6-methyladenosine methylation, RNA, Glioma, Neurogenesis
Address and Contact Information 1 Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
2 College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
3 Department of Acupuncture, Zaozhuang Traditional Chinese Medicine Hospital, Zaozhuang 277000, Shandong, China
4 First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250000, Shandong, China
5 Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
6 Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
7 Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
8 Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong, China
9 Suzhou Research Institute of Shandong University, Suzhou 215123, China
*Corresponding author: hanjinming1202@126.com; liyuan23@sdu.edu.cn
† Chunlin Li and Bowen Li have contributed equally to this work.
Read full article at BMC

No.  104DOI: 10.1186/s11658-023-00517-x Volume 28 (2023) - 28:104
Title MOLECULAR MECHANISM AND THERAPEUTIC POTENTIAL OF HDAC9 IN INTERVERTEBRAL DISC DEGENERATION
Authors Ming Lei1*†, Hui Lin1†, Deyao Shi1†, Pan Hong1, Hui Song1, Bomansaan Herman1, Zhiwei Liao1* and Cao Yang1*
Abstract Background: Intervertebral disc degeneration (IVDD) is the major cause of low-back pain. Histone deacetylase 9 (HDAC9) was dramatically decreased in the degenerative nucleus pulposus (NP) samples of patients with intervertebral disc degeneration (IVDD) according to bioinformatics analysis of Gene Expression Omnibus (GEO) GSE56081 dataset. This study aims to investigate the role of HDAC9 in IVDD progression.
Methods: The contribution of HDAC9 to the progression of IVDD was assessed using HDAC9 knockout (HDAC9KO) mice and NP-targeted HDAC9-overexpressing mice by IVD injection of adenovirus-mediated HDAC9 under a Col2a1 promoter. Magnetic resonance imaging (MRI) and histological analysis were used to examine the degeneration of IVD. NP cells were isolated from mice to investigate the effects of HDAC9 on apoptosis and viability. mRNA-seq and coimmunoprecipitation/mass spectrometry (co-IP/MS) analysis were used to analyze the HDAC9-regulated factors in the primary cultured NP cells.
Results: HDAC9 was statistically decreased in the NP tissues in aged mice. HDAC9KO mice spontaneously developed age-related IVDD compared with wild-type (HDAC9WT) mice. In addition, overexpression of HDAC9 in NP cells alleviated IVDD symptoms in a surgically-induced IVDD mouse model. In an in vitro assay, knockdown of HDAC9 inhibited cell viability and promoted cell apoptosis of NP cells, and HDAC9 overexpression had the opposite effects in NP cells isolated from HDAC9KO mice. Results of mRNA-seq and co-IP/MS analysis revealed the possible proteins and signaling pathways regulated by HDAC9 in NP cells. RUNX family transcription factor 3 (RUNX3) was screened out for further study, and RUNX3 was found to be deacetylated and stabilized by HDAC9. Knockdown of RUNX3 restored the effects of HDAC9 silencing on NP cells by inhibiting apoptosis and increasing viability.
Conclusion: Our results suggest that HDAC9 plays an important role in the development and progression of IVDD. It might be required to protect NP cells against the loss of cell viability and apoptosis by inhibiting RUNX3 acetylation and expression during IVDD. Together, our findings suggest that HDAC9 may be a potential therapeutic target in IVDD.
Keywords Intervertebral disc degeneration, HDAC9, RUNX3, Cell viability, Apoptosis
Address and Contact Information Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
*Corresponding author: leiming2010@hust.edu.cn; lzwhust@hust.edu.cn; caoyangunion@hust.edu.cn
Ming Lei, Hui Lin and Deyao Shi are contributed equally to this work.
Read full article at BMC

No.  105DOI: 10.1186/s11658-023-00518-w Volume 28 (2023) - 28:105
Title CircZNF609 REGULATES PULMONARY FIBROSIS VIA miR-145-5p/KLF4 AXIS AND ITS TRANSLATION FUNCTION
Authors Wenqing Sun1†, Siyun Zhou1†, Lan Peng1†, Yi Liu1, Demin Cheng1, Yue Wang1 and Chunhui Ni1,2*
Abstract Background: Pulmonary fibrosis is a growing clinical problem that develops as a result of abnormal wound healing, leading to breathlessness, pulmonary dysfunction and ultimately death. However, therapeutic options for pulmonary fibrosis are limited because the underlying pathogenesis remains incompletely understood. Circular RNAs, as key regulators in various diseases, remain poorly understood in pulmonary fibrosis induced by silica.
Methods: We performed studies with fibroblast cell lines and silica-induced mouse pulmonary fibrosis models. The expression of circZNF609, miR-145-5p, and KLF4 was determined by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RNA immunoprecipitation (RIP) assays and m6A RNA immunoprecipitation assays (MeRIP), Western blotting, immunofluorescence assays, and CCK8 were performed to investigate the role of the circZNF609/miR-145-5p/KLF4 axis and circZNF609-encoded peptides in fibroblast activation.
Results: Our data showed that circZNF609 was downregulated in activated fibroblasts and silica-induced fibrotic mouse lung tissues. Overexpression of circZNF609 could inhibit fibroblast activation induced by transforming growth factor-β1 (TGF-β1). Mechanically, we revealed that circZNF609 regulates pulmonary fibrosis via miR-145-5p/KLF4 axis and circZNF609-encoded peptides. Furthermore, circZNF609 was highly methylated and its expression was controlled by N6-methyladenosine (m6A) modification. Lastly, in vivo studies revealed that overexpression of circZNF609 attenuates silica-induced lung fibrosis in mice.
Conclusions: Our data indicate that circZNF609 is a critical regulator of fibroblast activation and silica-induced lung fibrosis. The circZNF609 and its derived peptides may represent novel promising targets for the treatment of pulmonary fibrosis.
Keywords Silicosis, circZNF609, Coding capacity, Non-coding RNA
Address and Contact Information 1 Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
2 Department of Public Health, Kangda College of Nanjing Medical University, Lianyungang 320700, China
*Corresponding author: chni@njmu.edu.cn; chninjmu@126.com
Wenqing Sun, Siyun Zhou and Lan Peng contributed equally to this work and should be considered co-first authors.
Read full article at BMC