Vol. 27 (2022)



No. 01 DOI: 10.1186/s11658-021-00301-9 Volume 27 (2022) - 27:01
Title THE ROLE OF POLYPHENOLS IN OVERCOMING CANCER DRUG RESISTANCE: A COMPREHENSIVE REVIEW
Authors Parisa Maleki Dana1, Fatemeh Sadoughi1, Zatollah Asemi1* and Bahman Yousef2,3*
Abstract Chemotherapeutic drugs are used to treat advanced stages of cancer or following surgery. However, cancers often develop resistance against drugs, leading to failure of treatment and recurrence of the disease. Polyphenols are a family of organic compounds with more than 10,000 members which have a three-membered favan ring system in common. These natural compounds are known for their benefcial properties, such as free radical scavenging, decreasing oxidative stress, and modulating infammation. Herein, we discuss the role of polyphenols (mainly curcumin, resveratrol, and epigallocatechin gallate [EGCG]) in diferent aspects of cancer drug resistance. Increasing drug uptake by tumor cells, decreasing drug metabolism by enzymes (e.g. cytochromes and glutathione-S-transferases), and reducing drug efux are some of the mechanisms by which polyphenols increase the sensitivity of cancer cells to chemotherapeutic agents. Polyphenols also afect other targets for overcoming chemoresistance in cancer cells, including cell death (i.e. autophagy and apoptosis), EMT, ROS, DNA repair processes, cancer stem cells, and epigenetics (e.g. miRNAs).
Keywords Polyphenols, Curcumin, Resveratrol, Epigallocatechin gallate, Chemoresistance
Address and Contact Information 1 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran
2 Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
3 Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.

*Corresponding author: asemi_z@Kaums.ac.ir; bahmanusef@gmail.com
Read full article at BMC

No. 02 DOI: 10.1186/s11658-021-00302-8 Volume 27 (2022) - 27:02
Title THE FUNCTIONS AND ROLES OF SESTRINS IN REGULATING HUMAN DISEASES
Authors Yitong Chen1†, Tingben Huang2†, Zhou Yu2, Qiong Yu2, Ying Wang3, Ji’an Hu4*, Jiejun Shi1* and Guoli Yang2*
Abstract Sestrins (Sesns), highly conserved stress-inducible metabolic proteins, are known to protect organisms against various noxious stimuli including DNA damage, oxidative stress, starvation, endoplasmic reticulum (ER) stress, and hypoxia. Sesns regulate metabolism mainly through activation of the key energy sensor AMP-dependent protein kinase (AMPK) and inhibition of mammalian target of rapamycin complex 1 (mTORC1). Sesns also play pivotal roles in autophagy activation and apoptosis inhibition in normal cells, while conversely promoting apoptosis in cancer cells. The functions of Sesns in diseases such as metabolic disorders, neurodegenerative diseases, cardiovascular diseases, and cancer have been broadly investigated in the past decades. However, there is a limited number of reviews that have summarized the functions of Sesns in the pathophysiological processes of human diseases, especially musculoskeletal system diseases. One aim of this review is to discuss the biological functions of Sesns in the pathophysiological process and phenotype of diseases. More signifcantly, we include some new evidence about the musculoskeletal system. Another purpose is to explore whether Sesns could be potential biomarkers or targets in the future diagnostic and therapeutic process.
Keywords Sestrins, Biological functions, Human diseases, Musculoskeletal system disease, Biomarker, Therapeutic target
Address and Contact Information 1 Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, Zhejiang, China
2 Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, Zhejiang, China
3 Department of Oral Medicine, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, Zhejiang,China.
4 Department of Oral Pathology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, Zhejiang, China
*Corresponding author: hja@zju.edu.cn; sjiejun@zju.edu.cn; 7308037@zju.edu.cn
Yitong Chen and Tingben Huang contributed equally to this work
Read full article at BMC

No. 03 DOI: 10.1186/s11658-021-00299-0 Volume 27 (2022) - 27:03
Title MITOCHONDRIAL POTASSIUM CHANNELS: A NOVEL CALCITRIOL TARGET
Authors Anna M. Olszewska1, Adam K. Sieradzan2, Piotr Bednarczyk3, Adam Szewczyk4 and Michał A. Żmijewski1*
Abstract Background: Calcitriol (an active metabolite of vitamin D) modulates the expression of hundreds of human genes by activation of the vitamin D nuclear receptor (VDR). However, VDR-mediated transcriptional modulation does not fully explain various phenotypic efects of calcitriol. Recently a fast non-genomic response to vitamin D has been described, and it seems that mitochondria are one of the targets of calcitriol. These non-classical calcitriol targets open up a new area of research with potential clinical applications. The goal of our study was to ascertain whether calcitriol can modulate mitochondrial function through regulation of the potassium channels present in the inner mitochondrial membrane.
Methods: The efects of calcitriol on the potassium ion current were measured using the patch-clamp method modifed for the inner mitochondrial membrane. Molecular docking experiments were conducted in the Autodock4 program. Additionally, changes in gene expression were investigated by qPCR, and transcription factor binding sites were analyzed in the CiiiDER program. Results: For the frst time, our results indicate that calcitriol directly afects the activity of the mitochondrial large-conductance Ca2+-regulated potassium channel (mitoBKCa) from the human astrocytoma (U-87 MG) cell line but not the mitochondrial calciumindependent two-pore domain potassium channel (mitoTASK-3) from human keratinocytes (HaCaT). The open probability of the mitoBKCa channel in high calcium conditions decreased after calcitriol treatment and the opposite efect was observed in low calcium conditions. Moreover, using the AutoDock4 program we predicted the binding poses of calcitriol to the calcium-bound BKCa channel and identifed amino acids interacting with the calcitriol molecule. Additionally, we found that calcitriol infuences the expression of genes encoding potassium channels. Such a dual, genomic and nongenomic action explains the pleiotropic activity of calcitriol. Conclusions: Calcitriol can regulate the mitochondrial large-conductance calcium regulated potassium channel. Our data open a new chapter in the study of nongenomic responses to vitamin D with potential implications for mitochondrial bioenergetics and cytoprotective mechanisms.
Keywords Calcitriol, Large-conductance calcium-regulated potassium channel, Mitochondria, Patch-clamp
Address and Contact Information 1 Department of Histology, Medical University of Gdańsk, 1a Dębinki, 80-211 Gdańsk, Poland
2 Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland.
3 Department of Physics and Biophysics, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
4 Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland.
*Corresponding author: mzmijewski@gumed.edu.pl
Read full article at BMC

No. 04 DOI: 10.1186/s11658-021-00300-w Volume 27 (2022) - 27:04
Title Specifcity of H2O2 signaling in leaf senescence: is the ratio of H2O2 contents in diferent cellular compartments sensed in Arabidopsis plants?
Authors Ulrike Zentgraf*, Ana Gabriela Andrade‐Galan and Stefan Bieker
Abstract Leaf senescence is an integral part of plant development and is driven by endogenous cues such as leaf or plant age. Developmental senescence aims to maximize the usage of carbon, nitrogen and mineral resources for growth and/or for the sake of the next generation. This requires efcient reallocation of the resources out of the senescing tissue into developing parts of the plant such as new leaves, fruits and seeds. However, premature senescence can be induced by severe and long-lasting biotic or abiotic stress conditions. It serves as an exit strategy to guarantee ofspring in an unfavorable environment but is often combined with a trade-of in seed number and quality. In order to coordinate the very complex process of developmental senescence with environmental signals, highly organized networks and regulatory cues have to be in place. Reactive oxygen species, especially hydrogen peroxide (H2O2), are involved in senescence as well as in stress signaling. Here, we want to summarize the role of H2O2 as a signaling molecule in leaf senescence and shed more light on how specifcity in signaling might be achieved. Altered hydrogen peroxide contents in specifc compartments revealed a diferential impact of H2O2 produced in diferent compartments. Arabidopsis lines with lower H2O2 levels in chloroplasts and cytoplasm point to the possibility that not the actual contents but the ratio between the two diferent compartments is sensed by the plant cells.
Keywords Leaf senescence, Free oxygen radicals, ROS, Hydrogen peroxide, Stromules, Senescence regulation, Intracellular compartments
Address and Contact Information ZMBP (Centre of Plant Molecular Biology), University of Tübingen, Auf der Morgenstelle 32, 72076 Tübingen, Germany
*Corresponding author: ulrike.zentgraf@zmbp.uni-tuebingen.de
Read full article at BMC

No. 05 DOI: 10.1186/s11658-021-00304-6 Volume 27 (2022) - 27:05
Title LncRNA HCG18 PROMOTES OSTEOSARCOMA GROWTH BY ENHANCED AEROBIC GLYCOLYSIS VIA THE miR‐365a‐3p/PGK1 AXIS
Authors Xiaohui Pan1, Jin Guo3, Canjun Liu4, Zhanpeng Pan1, Zhicheng Yang2*, Xiang Yao1* and Jishan Yuan1*
Abstract Background: Osteosarcoma (OS) is a common primary bone malignancy. Long non-coding RNA HCG18 is known to play an important role in a variety of cancers. However, its role in OS and relevant molecular mechanisms are unclear.
Methods: Real-time quantitative PCR was performed to determine the expression of target genes. Function experiments showed the efects of HCG18 and miR-365a-3p on OS cell growth.
Results: HCG18 expression was increased in OS cell lines. Moreover, in vitro and in vivo experiments demonstrated that HCG18 knockdown inhibited OS cell proliferation. Mechanistically, HCG18 was defned as a competing endogenous RNA by sponging miR-365a-3p, thus elevating phosphoglycerate kinase 1 (PGK1) expression by directly targeting its 3ʹUTR to increase aerobic glycolysis.
Conclusion: HCG18 promoted OS cell proliferation via enhancing aerobic glycolysis by regulating the miR-365a-3p/PGK1 axis. Therefore, HCG18 may be a potential target for OS treatment.
Keywords HCG18, miR-365a-3p, PGK1, Osteosarcoma, Aerobic glycolysis
Address and Contact Information 1 Department of Orthopedics, The Afliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, China
2 Department of Orthopedics, Changzhou No. 2 People’s Hospital, The Afliated Hospital of Nanjing Medical University, Changzhou, China
3 Department of Orthopedics, Zhenjiang First People’s Hospital Branch, Zhenjiang, People’s Republic of China.
4 Department of Respiratory Therapy, The Afliated People’s Hospital of Jiangsu University, Zhenjiang 212002, Jiangsu, China.
*Corresponding author: 727633793yzc@sina.com; yaoxiang6266@163.com; yuanjs2022@163.com
Read full article at BMC

No. 06 DOI: 10.1186/s11658-022-00308-w Volume 27 (2022) - 27:06
Title THE PROBABLE ROLE AND THERAPEUTIC POTENTIAL OF THE PI3K/AKT SIGNALING PATHWAY IN SARS‐CoV‐2 INDUCED COAGULOPATHY
Authors Mohammad Raf Khezri1*, Reza Varzandeh1 and Morteza Ghasemnejad‐Berenji1,2*
Abstract Coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is associated with a high mortality rate. The majority of deaths in this disease are caused by ARDS (acute respiratory distress syndrome) followed by cytokine storm and coagulation complications. Although alterations in the level of the number of coagulation factors have been detected in samples from COVID-19 patients, the direct molecular mechanism which has been involved in this pathologic process has not been explored yet. The PI3K/AKT signaling pathway is an intracellular pathway which plays a central role in cell survival. Also, in recent years the association between this pathway and coagulopathies has been well clarifed. Therefore, based on the evidence on over-activity of the PI3K/AKT signaling pathway in SARS-CoV-2 infection, in the current review, the probable role of this cellular pathway as a therapeutic target for the prevention of coagulation complications in patients with COVID-19 is discussed.
Keywords SARS-CoV-2, Coagulation, COVID-19, PI3K/AKT
Address and Contact Information 1 Department of Pharmacology and Toxicology, Faculty of Pharmacy, Urmia University of Medical Sciences, Sero Road, 5715799313 Urmia, Iran
2 Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran.
*Corresponding author: Drmnkh76@gmail.com; ghasemnejad.m@umsu.ac.ir
Read full article at BMC

No. 07 DOI: 10.1186/s11658-021-00305-5 Volume 27 (2022) - 27:07
Title THE INTERACTION OF CANONICAL Wnt/β‐CATENIN SIGNALING WITH PROTEIN LYSINE ACETYLATION
Authors Hongjuan You1†, Qi Li1,2†, Delong Kong1, Xiangye Liu1, Fanyun Kong1*, Kuiyang Zheng1,3 and Renxian Tang1,3*
Abstract Canonical Wnt/β-catenin signaling is a complex cell-communication mechanism that has a central role in the progression of various cancers. The cellular factors that participate in the regulation of this signaling are still not fully elucidated. Lysine acetylation is a signifcant protein modifcation which facilitates reversible regulation of the target protein function dependent on the activity of lysine acetyltransferases (KATs) and the catalytic function of lysine deacetylases (KDACs). Protein lysine acetylation has been classifed into histone acetylation and non-histone protein acetylation. Histone acetylation is a kind of epigenetic modifcation, and it can modulate the transcription of important biological molecules in Wnt/β-catenin signaling. Additionally, as a type of post-translational modifcation, non-histone acetylation directly alters the function of the core molecules in Wnt/β-catenin signaling. Conversely, this signaling can regulate the expression and function of target molecules based on histone or non-histone protein acetylation. To date, various inhibitors targeting KATs and KDACs have been discovered, and some of these inhibitors exert their anti-tumor activity via blocking Wnt/β-catenin signaling. Here, we discuss the available evidence in understanding the complicated interaction of protein lysine acetylation with Wnt/β-catenin signaling, and lysine acetylation as a new target for cancer therapy via controlling this signaling.
Keywords Protein lysine acetylation, Canonical Wnt/β-catenin signaling, Interaction, Therapy, Molecular mechanisms
Address and Contact Information 1 Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu, China.
2 Laboratory Department, The People’s Hospital of Funing, Yancheng, Jiangsu, China.
3 National Demonstration Center for Experimental Basic Medical Sciences Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
*Corresponding author: kong.fanyun@163.com; tangrenxian-t@163.com
Hongjuan You and Qi Li contributed equally to this work
Read full article at BMC

No. 08 DOI: 10.1186/s11658-022-00306-y Volume 27 (2022) - 27:08
Title CONSTRUCTION AND INVESTIGATION OF β3GNT2‐ASSOCIATED REGULATORY NETWORK IN ESOPHAGEAL CARCINOMA
Authors Zhiguo Luo1†, Qing Hu2†, Yuanhui Tang1, Yahui Leng2, Tian Tian2, Shuangyue Tian2, Chengyang Huang2, Ao Liu2, Xinzhou Deng1* and Li Shen1,2*
Abstract Background: Glycosyltransferases play a crucial role in various cancers. β1, 3-N-acetyl-glucosaminyltransferase 2, a polylactosamine synthase, is an important member of the glycosyltransferase family. However, the biological function and regulatory mechanism of β3GNT2 in esophageal carcinoma (ESCA) is still poorly understood.
Methods: The Cancer Genome Atlas and Genotype-Tissue Expression databases were used for gene expression and prognosis analysis. Quantitative real-time PCR, Western blot, and immunohistochemistry were performed to detect the expression of β3GNT2 in ESCA cell lines and tissues. In vitro assays and xenograft tumor models were utilized to evaluate the impact of β3GNT2 on ESCA progression. The downstream efectors and upstream regulators of β3GNT2 were predicted by online software and verifed by functional experiments.
Results: We found that β3GNT2 was highly expressed in ESCA tissues and positively correlated with poor prognosis in ESCA patients. β3GNT2 expression was closely associated with the tumor size, TNM stage, and overall survival of ESCA patients. Functionally, β3GNT2 promoted ESCA cell growth, migration, and invasion in vitro, as well as tumorigenesis in vivo. Mechanistically, β3GNT2 knockdown decreased the expression of the polylactosamine on EGFR. Knockdown of β3GNT2 also inhibited the JAK/STAT signaling pathway. Meanwhile, the JAK/STAT inhibitor could partly reverse the biological efects caused by β3GNT2 overexpression. Moreover, β3GNT2 expression was positively regulated by CREB1 and negatively regulated by miR-133b. Both CREB1 and miR-133b was involved in the β3GNT2-mediated ESCA progression.
Conclusions: Our study, for the frst time, reveals the importance of β3GNT2 in ESCA progression and ofers a potential therapeutic target for ESCA.
Keywords Esophageal carcinoma, Progression, Glycosyltransferase, β3GNT2
Address and Contact Information 1 Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China
2 Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.
*Corresponding author: 576700586@qq.com; 20101061@hbmu.edu.cn
Zhiguo Luo and Qing Hu contributed equally to this article
Read full article at BMC

No. 09 DOI: 10.1186/s11658-022-00307-x Volume 27 (2022) - 27:09
Title RAMAN SPECTROSCOPY BIOCHEMICAL CHARACTERISATION OF BLADDER CANCER CISPLATIN RESISTANCE REGULATED BY FDFT1: A REVIEW
Authors M. Kanmalar1, Siti Fairus Abdul Sani1*, Nur Izzahtul Nabilla B. Kamri1, Nur Akmarina B. M. Said2, Amirah Hajirah B. A. Jamil2, S. Kuppusamy3, K. S. Mun4 and D. A. Bradley5,6
Abstract Bladder cancer is the fourth most common malignancy in males. It can present across the whole continuum of severity, from mild through well-diferentiated disease to extremely malignant tumours with poor survival rates. As with other vital organ malignancies, proper clinical management involves accurate diagnosis and staging. Chemotherapy consisting of a cisplatin-based regimen is the mainstay in the management of muscle-invasive bladder cancers. Control via cisplatin-based chemotherapy is threatened by the development of chemoresistance. Intracellular cholesterol biosynthesis in bladder cancer cells is considered a contributory factor in determining the chemotherapy response. Farnesyl-diphosphate farnesyltransferase 1 (FDFT1), one of the main regulatory components in cholesterol biosynthesis, may play a role in determining sensitivity towards chemotherapy compounds in bladder cancer. FDFT1-associated molecular identifcation might serve as an alternative or appendage strategy for early prediction of potentially chemoresistant muscle-invasive bladder cancer tissues. This can be accomplished using Raman spectroscopy. Developments in the instrumentation have led to it becoming one of the most convenient forms of analysis, and there is a highly realistic chance that it will become an efective tool in the pathology lab. Chemosensitive bladder cancer tissues tend to have a higher lipid content, more protein genes and more cholesterol metabolites. These are believed to be associated with resistance towards bladder cancer chemotherapy. Herein, Raman peak assignments have been tabulated as an aid to indicating metabolic changes in bladder cancer tissues that are potentially correlated with FDFT1 expression.
Keywords Bladder cancer, Diagnostic, FDFT1, Cisplatin chemoresistance, Raman spectroscopy
Address and Contact Information 1 Department of Physics, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
2 Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, 50603 Kuala Lumpur, Malaysia.
3 Department of Surgery, University of Malaya, 50603 Kuala Lumpur, Malaysia.
4 Department of Pathology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
5 Centre for Applied Physics and Radiation Technologies, Sunway University, Jalan University, 46150 Petaling Jaya, Malaysia.
6 Department of Physics, University of Surrey, Guildford GU2 7XH, UK.
*Corresponding author: s.fairus@um.edu.my
Read full article at BMC

No. 10 DOI: 10.1186/s11658-022-00311-1 Volume 27 (2022) - 27:10
Title THE ROLES OF Eph RECEPTORS, NEUROPILIN‐1, P2X7, AND CD147 IN COVID‐19‐ASSOCIATED NEURODEGENERATIVE DISEASES: INFAMMASOME AND JaK INHIBITORS AS POTENTIAL PROMISING THERAPIES
Authors Hamidreza Zalpoor1,2,3†, Abdullatif Akbari4†, Azam Samei5, Razieh Forghaniesfdvajani4, Monireh Kamali6, Azadeh Afzalnia6, Shirin Manshouri6, Fatemeh Heidari7, Majid Pornour8, Majid Khoshmirsafa9, Hossein Aazami10 and Farhad Seif2,11*
Abstract The novel coronavirus disease 2019 (COVID-19) pandemic has spread worldwide, and fnding a safe therapeutic strategy and efective vaccine is critical to overcoming severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, elucidation of pathogenesis mechanisms, especially entry routes of SARS-CoV-2 may help propose antiviral drugs and novel vaccines. Several receptors have been demonstrated for the interaction of spike (S) protein of SARS-CoV-2 with host cells, including angiotensin-converting enzyme (ACE2), ephrin ligands and Eph receptors, neuropilin 1 (NRP-1), P2X7, and CD147. The expression of these entry receptors in the central nervous system (CNS) may make the CNS prone to SARS-CoV-2 invasion, leading to neurodegenerative diseases. The present review provides potential pathological mechanisms of SARS-CoV-2 infection in the CNS, including entry receptors and cytokines involved in neuroinfammatory conditions. Moreover, it explains several neurodegenerative disorders associated with COVID-19. Finally, we suggest infammasome and JaK inhibitors as potential therapeutic strategies for neurodegenerative diseases.
Keywords COVID-19, CNS, Ephrin, Neuropilin-1, P2X7, CD147, Cytokine, Jak, Infammasome, Neurodegenerative diseases, Alzheimer’s disease, Parkinson’s disease
Address and Contact Information 1 American Association of Kidney Patients, Tampa, FL, USA.
2 Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran.
3 Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
4 Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientifc Education and Research Network (USERN), Tehran, Iran.
5 Department of Laboratory Sciences, School of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
6 Rajaei Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
7 Immunology Department, Faculty of Medicine, Tarbiat Modares University, Tehran, Iran.
8 Department of Oncology, School of Medicine, University of Maryland, Maryland, USA.
9 Department of Immunology, School of Medicine, Iran University of Medical Sciences, Iran, Iran.
10 Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
11 Department of Immunology and Allergy, Academic Center for Education, Culture, and Research (ACECR), Enghelab St., Aboureyhan St., Vahid Nazari Crossroad, P17, 1315795613 Tehran, Iran
*Corresponding author: farhad.seif@outlook.com
† Hamidreza Zalpoor and Abdullatif Akbari contributed equally as the first authors
Read full article at BMC

No. 11 DOI: 10.1186/s11658-022-00314-y Volume 27 (2022) - 27:11
Title Slc25a5 REGULATES ADIPOGENESIS BY MODULATING ERK SIGNALING IN OP9 CELLS
Authors Shenglong Zhu1,2†, Wei Wang1†, Jingwei Zhang3, Siyu Ji1, Zhe Jing1 and Yong Q. Chen1,2,3*
Abstract Background: A comprehensive understanding of the molecular mechanisms of adipogenesis is a critically important strategy for identifying new targets for obesity intervention.
Methods: Transcriptomic and lipidomic approaches were used to explore the functional genes regulating adipogenic diferentiation and their potential mechanism of action in OP9 cells and adipose-derived stem cells. Oil Red O staining was used to detect oil droplets in adipocytes.
Results: RNA sequencing (RNA-seq) showed that Slc25a5 expression was signifcantly upregulated in adipogenic diferentiation. Depletion of Slc25a5 led to the suppressed expression of adipogenesis-related genes, reduced the accumulation of triglycerides, and inhibited PPARγ protein expression. Moreover, the knockdown of Slc25a5 resulted in signifcant reduction of oxidative phosphorylation (OXPHOS) protein expression (ATP5A1, CQCRC2, and MTCO1) and ATP production. The RNA-seq and real-time quantitative polymerase chain reaction (RT–qPCR) results suggested that adipogenic diferentiation is possibly mediated by ERK1/2 phosphorylation, and this hypothesis was confrmed by intervention with PD98059 (an ERK 1/2 inhibitor).
Conclusions: This study indicates that Slc25a5 inhibits adipogenesis and might be a new therapeutic target for the treatment of obesity.
Keywords Obesity, Adipogenic diferentiation, Slc25a5, ERK, Transcriptome, Metabolome
Address and Contact Information 1 Wuxi School of Medicine, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, China
2 Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China.
3 School of Food Science and Technology, Jiangnan University, Wuxi, China.
*Corresponding author: yqc_lab@126.com; yqchen@jiangnan.edu.cn
Shenglong Zhu and Wei Wang contributed equally to this work
Read full article at BMC

No. 12 DOI: 10.1186/s11658-022-00315-x Volume 27 (2022) - 27:12
Title ERp57/PDIA3: NEW INSIGHT
Authors Silvia Chichiarelli1*, Fabio Altieri1, Giuliano Paglia1, Elisabetta Rubini1,2, Marco Minacori1 and Margherita Eufemi1
Abstract The ERp57/PDIA3 protein is a pleiotropic member of the PDIs family and, although predominantly located in the endoplasmic reticulum (ER), has indeed been found in other cellular compartments, such as the nucleus or the cell membrane. ERp57/PDIA3 is an important research target considering it can be found in various subcellular locations. This protein is involved in many diferent physiological and pathological processes, and our review describes new data on its functions and summarizes some ligands identifed as PDIA3-specifc inhibitors.
Keywords ERp57, PDIA3, PDI inhibitors, Punicalagin, Vitamin D3, Cancer, Infections, Nervous system, Cardiovascular system, Fertility
Address and Contact Information 1 Department of Biochemical Sciences “A.Rossi-Fanelli”, Sapienza University of Rome, P.le A.Moro 5, 00185 Rome, Italy
2 Enrico Ed Enrica Sovena” Foundation, Rome, Italy.
*Corresponding author: silvia.chichiarelli@uniroma1.it
Read full article at BMC

No. 13 DOI: 10.1186/s11658-022-00313-z Volume 27 (2022) - 27:
Title SNHG3 COULD PROMOTE PROSTATE CANCER PROGRESSION THROUGH REDUCING METHIONINE DEPENDENCE OF PCa CELLS
Authors Xiaotian Wang, Yongsheng Song, Yaxing Shi, Da Yang, Jiaxing Li and Bo Yin*
Abstract In recent years, morbidity and mortality of prostate cancer (PCa) have increased dramatically, while mechanistic understanding of its onset and progression remains unmet. LncRNA SNHG3 has been proved to stimulate malignant progression of multiple cancers, whereas its functional mechanism in PCa needs to be deciphered. In this study, our analysis in the TCGA database revealed high SNHG3 expression in PCa tissue. Further analysis in starBase, TargetScan, and mirDIP databases identifed the SNHG3/miR-152-3p/SLC7A11 regulatory axis. FISH was conducted to assess the distribution of SNHG3 in PCa tissue. Dual-luciferase reporter gene and RIP assays confrmed the relationship among the three objects. Next, qRT-PCR and western blot were conducted to measure expression levels of SNHG3, miR-152-3p, and SLC7A11. CCK-8, colony formation, Transwell, and fow cytometry were carried out to assess proliferation, migration, invasion, methionine dependence, apoptosis, and the cell cycle. It was noted that SNHG3 as a molecular sponge of miR-152-3p stimulated proliferation, migration, and invasion, restrained methionine dependence and apoptosis, and afected the cell cycle of PCa cells via targeting SLC7A11. Additionally, we constructed xenograft tumor models in nude mice and confrmed that knockdown of SNHG3 could restrain PCa tumor growth and elevate methionine dependence in vivo. In conclusion, our investigation improved understanding of the molecular mechanism of SNHG3 modulating PCa progression, thereby generating novel insights into clinical therapy for PCa.
Keywords Prostate cancer, SNHG3, miR-152-3p, SLC7A11, Methionine dependence
Address and Contact Information Department of Urology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang 110001, Liaoning, China
*Corresponding author: yinbo19751003@hotmail.com
Read full article at BMC

No. 14 DOI: 10.1186/s11658-022-00317-9 Volume 27 (2022) - 27:14
Title MicroRNA let‐7 AND VIRAL INFECTIONS: FOCUS ON MECHANISMS OF ACTION
Authors Arash Letafati1, Sajad Najaf2, Mehran Mottahedi3, Mohammad Karimzadeh4, Ali Shahini3, Setareh Garousi3, Mohammad Abbasi‐Kolli5, Javid Sadri Nahand6, Seyed Saeed Tamehri Zadeh7, Michael R. Hamblin8, Neda Rahimian9,10*, Mohammad Taghizadieh11* and Hamed Mirzaei12,13*
Abstract MicroRNAs (miRNAs) are fundamental post-transcriptional modulators of several critical cellular processes, a number of which are involved in host defense mechanisms. In particular, miRNA let-7 functions as an essential regulator of the function and diferentiation of both innate and adaptive immune cells. Let-7 is involved in several human diseases, including cancer and viral infections. Several viral infections have found ways to dysregulate the expression of miRNAs. Extracellular vesicles (EV) are membrane-bound lipid structures released from many types of human cells that can transport proteins, lipids, mRNAs, and miRNAs, including let-7. After their release, EVs are taken up by the recipient cells and their contents released into the cytoplasm. Let-7-loaded EVs have been suggested to afect cellular pathways and biological targets in the recipient cells, and can modulate viral replication, the host antiviral response, and the action of cancer-related viruses. In the present review, we summarize the available knowledge concerning the expression of let-7 family members, functions, target genes, and mechanistic involvement in viral pathogenesis and host defense. This may provide insight into the development of new therapeutic strategies to manage viral infections.
Keywords MicroRNAs, Let-7, Viral infections, Regulatory role
Address and Contact Information 1 Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
2 Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
3 Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
4 Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
5 Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
6 Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
7 School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
8 Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa.
9 Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
10 Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
11 Department of Pathology, School of Medicine, Center for Women’s Health Research Zahra, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran.
12 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran. 13Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
*Corresponding author: rahimian.n@iums.ac.ir; MohammadTaghizadieh@gmail.com; mirzaei-h@kaums.ac.ir; h.mirzaei2002@gmail.com
Read full article at BMC

No. 15 DOI: 10.1186/s11658-022-00310-2 Volume 27 (2022) - 27:15
Title THE lncRNA DANCR PROMOTES DEVELOPMENT OF ATHEROSCLEROSIS BY REGULATING THE miR‐214‐5p/COX20 SIGNALING PATHWAY
Authors Ruolan Zhang1*, Yuming Hao2 and Jinrong Zhang1
Abstract Background: Although long non-coding RNA diferentiation antagonizing non-protein coding RNA (DANCR) has been reported to be involved in atherosclerosis (AS) development, its specifc mechanism remains unclear.
Methods: DANCR expression levels in blood samples of AS patients and oxidized low-density lipoprotein (ox-LDL) treated vascular smooth muscle cells (VSMCs) and human umbilical vein endothelial cells (HUVECs) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). The small interfering RNA targeting DANCR (si-DANCR) was used to silence DANCR expression. Cell viability was assessed by CCK-8 assay. Cell apoptosis was evaluated by fow cytometry. Levels of infammatory cytokines, anti-oxidative enzyme superoxide dismutase (SOD) activity, and malonaldehyde (MDA) were detected by specifc commercial kits. An animal AS model was established to confrm the role of DANCR/microR-214-5p/COX20 (the chaperone of cytochrome c oxidase subunit II COX2) in AS development.
Results: DANCR was signifcantly increased in the blood samples of AS patients and ox-LDL treated VSMCs and HUVECs. DANCR downregulation obviously increased viability and reduced apoptosis of ox-LDL-treated VSMCs and HUVECs. Meanwhile, DANCR downregulation reduced the levels of infammatory cytokines, including interleukin (IL)-6 (IL-6), IL-1beta (IL-1β), IL-6 and tumor necrosis factor (TNF)-alpha (TNF-α) and MDA while increasing the SOD level in ox-LDL-treated VSMCs and HUVECs. DANCR regulated COX20 expression by acting as a competing endogenous RNA (ceRNA) of miR-214-5p. Rescue experiments demonstrated that miR-214-5p downregulation obviously attenuated si-DANCR-induced protective efects on ox-LDL-caused endothelial injury.
Conclusions: Our results revealed that DANCR promoted AS progression by targeting the miR-214-5p/COX20 axis, suggesting that DANCR might be a potential therapeutic target for AS.
Keywords Atherosclerosis, DANCR, miR-214-5p, COX20
Address and Contact Information 1 Department of Cardiology, Harrison International Peace Hospital, No. 180 Renmin Road, Hengshui City 053000, Hebei Province, People’s Republic of China
2 Department of Cardiology, Second Afliated Hospital of Hebei Medical University, Shijiazhuang City 05000, Hebei Province, People’s Republic of China.
*Corresponding author: ruolanzhangharriso@163.com
Read full article at BMC

No. 16 DOI: 10.1186/s11658-022-00316-w Volume 27 (2022) - 27:16
Title EXPLORING NEURONAL MECHANISMS INVOLVED IN THE SCRATCHING BEHAVIOR OF A MOUSE MODEL OF ALLERGIC CONTACT DERMATITIS BY TRANSCRIPTOMICS
Authors Boyu Liu1†, Ruixiang Chen1,2†, Jie Wang1†, Yuanyuan Li1†, Chengyu Yin1, Yan Tai3, Huimin Nie1, Danyi Zeng1, Junfan Fang1, Junying Du1, Yi Liang1, Xiaomei Shao1, Jianqiao Fang1* and Boyi Liu1*
Abstract Background: Allergic contact dermatitis (ACD) is a common skin condition characterized by contact hypersensitivity to allergens, accompanied with skin infammation and a mixed itch and pain sensation. The itch and pain dramatically afects patients’ quality of life. However, still little is known about the mechanisms triggering pain and itch sensations in ACD.
Methods: We established a mouse model of ACD by sensitization and repetitive challenge with the hapten oxazolone. Skin pathological analysis, transcriptome RNA sequencing (RNA-seq), qPCR, Ca2+ imaging, immunostaining, and behavioral assay were used for identifying gene expression changes in dorsal root ganglion innervating the infamed skin of ACD model mice and for further functional validations.
Results: The model mice developed typical ACD symptoms, including skin dryness, erythema, excoriation, edema, epidermal hyperplasia, infammatory cell infltration, and scratching behavior, accompanied with development of eczematous lesions. Transcriptome RNA-seq revealed a number of diferentially expressed genes (DEGs), including 1436-DEG mRNAs and 374-DEG-long noncoding RNAs (lncRNAs). We identifed a number of DEGs specifcally related to sensory neuron signal transduction, pain, itch, and neuroinfammation. Comparison of our dataset with another published dataset of atopic dermatitis mouse model identifed a core set of genes in peripheral sensory neurons that are exclusively afected by local skin infammation. We further found that the expression of the pain and itch receptor MrgprD was functionally upregulated in dorsal root ganglia (DRG) neurons innervating the infamed skin of ACD model mice. MrgprD activation induced by its agonist β-alanine resulted in exaggerated scratching responses in ACD model mice compared with naïve mice.
Conclusions: We identifed the molecular changes and cellular pathways in peripheral sensory ganglia during ACD that might participate in neurogenic infammation, pain, and itch. We further revealed that the pain and itch receptor MrgprD is functionally upregulated in DRG neurons, which might contribute to peripheral pain and itch sensitization during ACD. Thus, targeting MrgprD may be an efective method for alleviating itch and pain in ACD.
Keywords Itch, Pain, Sensory neurons, Allergic contact dermatitis, RNA-seq
Address and Contact Information 1 Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China.
2 The First Department of Acupuncture, Shaanxi Hospital of Traditional Chinese Medicine, Xi’an, Shaanxi, China.
3 Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
*Corresponding author: fangjianqiao7532@163.com; boyi.liu@foxmail.com
Boyu Liu, Ruixiang Chen, Jie Wang and Yuanyuan Li contributed equally to this work
Read full article at BMC

No. 17 DOI: 10.1186/s11658-022-00309-9 Volume 27 (2022) - 27:17
Title EXOSOMAL lncRNA TUG1 FROM CANCER‐ASSOCIATED FBROBLASTS PROMOTES LIVER CANCER CELL MIGRATION, INVASION, AND GLYCOLYSIS BY REGULATING THE miR‐524‐5p/SIX1 AXIS
Authors Le Lu, Jingjing Huang, Jiantao Mo, Xuanbo Da, Qiaoxin Li, Meng Fan and Hongwei Lu*
Abstract Background: Increasing evidence suggests that taurine upregulated gene 1 (TUG1) is crucial for tumor progression; however, its role in hepatocellular carcinoma (HCC) and the underlying mechanisms are not well characterized.
Methods: The expression levels of TUG1, miR-524-5p, and sine oculis homeobox homolog 1 (SIX1) were determined using quantitative real-time PCR. The regulatory relationships were confrmed by dual-luciferase reporter assay. Cell proliferation and invasion were assessed using Cell Counting Kit 8 and transwell assays. Glucose uptake, cellular levels of lactate, lactate dehydrogenase (LDH), and adenosine triphosphate (ATP) were detected using commercially available kits. Silencing of TUG1 or SIX1 was performed by lentivirus transduction. Protein levels were measured by immunoblotting.
Results: Cancer-associated fbroblasts (CAFs)-secreted exosomes promoted migration, invasion, and glycolysis in HepG2 cells by releasing TUG1. The promotive efects of CAFs-secreted exosomes were attenuated by silencing of TUG1. TUG1 and SIX1 are targets of miR-524-5p. SIX1 knockdown inhibited the promotive efects of miR-524-5p inhibitor. Silencing of TUG1 suppressed tumor growth and lung metastasis and therefore increased survival of xenograft model mice. We also found that TUG1 and SIX1 were increased in HCC patients with metastasis while miR-524-5p was decreased in HCC patients with metastasis.
Conclusions: CAFs-derived exosomal TUG1 promoted migration, invasion, and glycolysis in HCC cells via the miR-524-5p/SIX1 axis. These fndings may help establish the foundation for the development of therapeutics strategies and clinical management for HCC in future.
Keywords Long noncoding RNA, Taurine upregulated gene 1, Hepatocellular carcinoma, microRNA, Sine oculis homeobox homolog 1, Exosomes
Address and Contact Information Department of General Surgery, The Second Afliated Hospital of Xi’an Jiaotong University, No.157, West 5th Road, Xi’an 710004, China
*Corresponding author: lhwdoc@163.com; lhwlhw135@163.com
Read full article at BMC

No. 18 DOI: 10.1186/s11658-022-00323-x Volume 27 (2022) - 27:18
Title IDENTIFYING PATHWAYS REGULATING THE ONCOGENIC p53 FAMILY MEMBER ΔNp63 PROVIDES THERAPEUTIC AVENUES FOR SQUAMOUS CELL CARCINOMA
Authors Zuzana Pokorna, Jan Vyslouzil, Borivoj Vojtesek and Philip J. Coates*
Abstract Background: ΔNp63 overexpression is a common event in squamous cell carcinoma (SCC) that contributes to tumorigenesis, making ΔNp63 a potential target for therapy.
Methods: We created inducible TP63-shRNA cells to study the efects of p63-depletion in SCC cell lines and non-malignant HaCaT keratinocytes. DNA damaging agents, growth factors, signaling pathway inhibitors, histone deacetylase inhibitors, and metabolism-modifying drugs were also investigated for their ability to infuence ΔNp63 protein and mRNA levels.
Results: HaCaT keratinocytes, FaDu and SCC-25 cells express high levels of ΔNp63. HaCaT and FaDu inducible TP63-shRNA cells showed reduced proliferation after p63 depletion, with greater efects on FaDu than HaCaT cells, compatible with oncogene addiction in SCC. Genotoxic insults and histone deacetylase inhibitors variably reduced ΔNp63 levels in keratinocytes and SCC cells. Growth factors that regulate proliferation/survival of squamous cells (IGF-1, EGF, amphiregulin, KGF, and HGF) and PI3K, mTOR, MAPK/ERK or EGFR inhibitors showed lesser and inconsistent efects, with dual inhibition of PI3K and mTOR or EGFR inhibition selectively reducing ΔNp63 levels in HaCaT cells. In contrast, the antihyperlipidemic drug lovastatin selectively increased ΔNp63 in HaCaT cells.
Conclusions: These data confrm that ΔNp63-positive SCC cells require p63 for continued growth and provide proof of concept that p63 reduction is a therapeutic option for these tumors. Investigations of ΔNp63 regulation identifed agent-specifc and cell-specifc pathways. In particular, dual inhibition of the PI3K and mTOR pathways reduced ΔNp63 more efectively than single pathway inhibition, and broad-spectrum histone deacetylase inhibitors showed a time-dependent biphasic response, with high level downregulation at the transcriptional level within 24 h. In addition to furthering our understanding of ΔNp63 regulation in squamous cells, these data identify novel drug combinations that may be useful for p63-based therapy of SCC.
Keywords ΔNp63, Oncogene addiction, Squamous cell carcinoma, DNA damage, Histone deacetylase inhibitors, Growth factor signaling
Address and Contact Information Research Center of Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, Zluty kopec 7, 656 53 Brno, Czech Republic
*Corresponding author: philip.coates@mou.cz
Read full article at BMC

No. 19 DOI: 10.1186/s11658-022-00312-0 Volume 27 (2022) - 27:19
Title PEPTIDYLARGININE DEIMINASE 2 PROMOTES T HELPER 17‐LIKE T CELL ACTIVATION AND ACTIVATED T CELL‐AUTONOMOUS DEATH (ACAD) THROUGH AN ENDOPLASMIC RETICULUM STRESS AND AUTOPHAGY COUPLING MECHANISM
Authors Yi‐Fang Yang1,2†, Chuang‐Ming Wang3†, I.‐Hsin Hsiao1, Yi‐Liang Liu1,4, Wen‐Hao Lin1,4, Chih‐Li Lin4, Hui‐Chih Hung1,6,7* and Guang‐Yaw Liu4,5*
Abstract Peptididylarginine deiminase type 2 (PADI2) catalyzes the conversion of arginine residues to citrulline residues on proteins. We demonstrate that PADI2 induces T cell activation and investigate how PADI2 promotes activated T cell autonomous death (ACAD). In activated Jurkat T cells, overexpression of PADI2 signifcantly increases citrullinated proteins and induces endoplasmic reticulum (ER) stress and unfolded protein response (UPR) signaling, ultimately resulting in the expression of autophagy-related proteins and autophagy. PADI2 promoted autophagy and resulted in the early degradation of p62 and the light chain 3B (LC3B)-II accumulation. In Jurkat T cells, silencing the autophagy-related gene (Atg) 12 protein inhibits PADI2-mediated autophagy and promotes ER stress and apoptosis, whereas overexpression of Atg12 decreased ER stress and prolonged autophagy to promote cell survival. Additionally, PADI2 regulates T cell activation and the production of Th17 cytokines in Jurkat T cells (interleukins 6, IL-17A, IL-17F, IL-21, and IL-22). In Jurkat T cells, silencing IL-6 promotes autophagy mediated by PADI2 and inhibits PADI2-induced apoptosis, whereas silencing Beclin-1 increases the activation and survival of Th17-like T cells while decreasing autophagy and apoptosis. PADI2 silencing alleviates ER stress caused by PADI2 and decreases cytokine expression associated with Th17-like T cell activation and ACAD. We propose that PADI2 was involved in Th17 lymphocyte ACAD via a mechanism involving ER stress and autophagy that was tightly regulated by PADI2-mediated citrullination. These fndings suggest that inhibiting Th17 T cell activation and the development of severe autoimmune diseases may be possible through the use of novel antagonists that specifcally target PADI2.
Keywords Peptidylarginine deiminase 2, Cytokines, Activated T cell-autonomous death, Endoplasmic reticulum stress, Autophagy
Address and Contact Information 1 Department of Life Sciences, National Chung Hsing University (NCHU), Taichung 40227, Taiwan
2 Ph.D. Program in Tissue Engineering and Regenerative Medicine, National Chung Hsing University, Taichung 40227, Taiwan.
3 Department of Pediatrics, Ditmanson Medical Foundation Chia-Yi Christian Hospital (CYCH), Chia‐Yi 60002, Taiwan.
4 Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan.
5 Department of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan.
6 Institute of Genomics and Bioinformatics, National Chung Hsing University (NCHU), Taichung 40227, Taiwan.
7 iEGG and Animal Biotechnology Center, NCHU, Taichung 40227, Taiwan.
*Corresponding author: hchung@dragon.nchu.edu.tw; liugy@csmu.edu.tw
Yi-Fang Yang and Chuang Ming Wang contributed equally to this work
Read full article at BMC

No. 20 DOI: 10.1186/s11658-022-00319-7 Volume 27 (2022) - 27:20
Title SENESCENCE‐ASSOCIATED REPROGRAMMING INDUCED BY INTERLEUKIN‐1 IMPAIRS RESPONSE TO EGFR NEUTRALIZATION
Authors Donatella Romaniello1,2†, Valerio Gelfo1,2†, Federica Pagano1, Enea Ferlizza1, Michela Sgarzi1,2, Martina Mazzeschi1,2, Alessandra Morselli1, Carmen Miano3, Gabriele D’Uva1,3 and Mattia Lauriola1,2*
Abstract Background: EGFR targeting is currently the main treatment strategy for metastatic colorectal cancer (mCRC). Results of diferent clinical trials show that patients with wild-type KRAS and BRAF beneft from anti-EGFR monoclonal antibodies (moAbs) cetuximab (CTX) or panitumumab. Unfortunately, despite initial response, patients soon became refractory. Tumor heterogeneity and multiple escaping routes have been addressed as the main culprit, and, behind genomic alterations already described, changes in signaling pathways induced by drug pressure are emerging as mechanisms of acquired resistance. We previously reported an association between reduced sensitivity to CTX and increased expression of IL-1. However, how IL-1 mediates CTX resistance in mCRC is still unclear.
Methods: Under CTX treatment, the upregulation of IL-1R1 expression and a senescence program in sensitive colorectal cancer (CRC) cell lines is examined over time using qPCR, immunoblotting, and immunofuorescence.
Results: In sensitive CRC cells, IL-1 appeared responsible for a CTX-mediated G0 phase arrest. On the contrary, CTX-resistant CRC cells (CXR) maintained high mRNA levels of IL-1R1 and a post-senescence reprogramming, as indicated by increased SNAIL expression. Interestingly, treatment of CXR cells with a recombinant decoy, able to sequester the soluble form of IL-1, pushed CTX-resistant CRC cells back into a stage of senescence, thus blocking their proliferation. Our model suggests a trans-regulatory mechanism mediated by IL-1 on EGFR signaling. By establishing senescence and regulating EGFR activity and expression, IL-1 exposure ultimately bestows resistance.
Conclusions: To sum up, our fndings point to the combined blockage of IL-1R and EGFR as a promising therapeutical approach to restore sensitivity to EGFR-targeting monoclonal antibodies.
Keywords EGFR, Pseudo-senescence, IL-1, Cell plasticity, Colon cancer, Cetuximab, Resistance
Address and Contact Information 1 Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy
2 Centre for Applied Biomedical Research (CRBA), Bologna University Hospital Authority St. Orsola-Malpighi Polyclinic, 40138 Bologna, Italy.
3 National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy.
*Corresponding author: mattia.lauriola2@unibo.it
Donatella Romaniello and Valerio Gelfo contributed equally to this work.
Read full article at BMC

No. 21 DOI: 10.1186/s11658-022-00320-0 Volume 27 (2022) - 27:21
Title POTENTIAL ANTICANCER PROPERTIES AND MECHANISMS OF THYMOQUINONE IN OSTEOSARCOMA AND BONE METASTASIS
Authors Mina Homayoonfal1, Zatollah Asemi1* and Bahman Yousef2,3*
Abstract Despite great advances, therapeutic approaches of osteosarcoma, the most prevalent class of preliminary pediatric bone tumors, as well as bone-related malignancies, continue to demonstrate insufcient adequacy. In recent years, a growing trend toward applying natural bioactive compounds, particularly phytochemicals, as novel agents for cancer treatment has been observed. Bioactive phytochemicals exert their anticancer features through two main ways: they induce cytotoxic efects against cancerous cells without having any detrimental impact on normal cell macromolecules such as DNA and enzymes, while at the same time combating the oncogenic signaling axis activated in tumor cells. Thymoquinone (TQ), the most abundant bioactive compound of Nigella sativa, has received considerable attention in cancer treatment owing to its distinctive properties, including apoptosis induction, cell cycle arrest, angiogenesis and metastasis inhibition, and reactive oxygen species (ROS) generation, along with inducing immune system responses and reducing side efects of traditional chemotherapeutic drugs. The present review is focused on the characteristics and mechanisms by which TQ exerts its cytotoxic efects on bone malignancies.
Keywords Osteosarcoma, Bone metastasis, Thymoquinone, Signaling pathway, Apoptosis, Angiogenesis, Chemotherapy resistance
Address and Contact Information 1 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
2 Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
3 Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
*Corresponding author: Asemi_r@yahoo.com; bahmanusef@gmail.com
Read full article at BMC

No. 22 DOI: 10.1186/s11658-022-00322-y Volume 27 (2022) - 27:22
Title miR‐152‐3p IMPEDES THE MALIGNANT PHENOTYPES OF HEPATOCELLULAR CARCINOMA BY REPRESSING ROUNDABOUT GUIDANCE RECEPTOR 1
Authors Tao Yin* and Haonan Zhao
Abstract Background: miR-152-3p functions as a tumour suppressor in the progression of hepatic tumorigenesis. Herein, we further discussed the prognostic signifcance and immune infltration of miR-152-3p and its potential gene target in hepatocellular carcinoma (HCC).
Methods: The Cancer Genome Atlas (TCGA), Integrative Molecular Database of Hepatocellular Carcinoma (HCCDB), Human Protein Atlas (HPA) and Kaplan–Meier Plotter databases were used to evaluate miR-152-3p and roundabout guidance receptor 1 (ROBO1) expression, prognosis and immune infltration. In vitro cell experiments, including cell proliferation and apoptosis, were evaluated using Cell Counting Kit 8 (CCK8) and terminal-deoxynucleotidyl transferase-mediated nick end labelling (TUNEL) assays.
Results: Up-regulation of ROBO1 functioned as an oncogene associated with poor prognosis, immune cell enrichment and cell proliferation in HCC. ROBO1 was signifcantly positively correlated with the enrichment of multiple immune cells and their biomarkers. Enrichment of type-2 T-helper (Th2) cells is an unfavourable biomarker of HCC prognosis. GSEA revealed that ROBO1 correlated with apoptosis, mitosis and carcinogenic signalling pathways. Suppression of cell proliferation and the enhancement of cell apoptosis by miR-152-3p mimics were counteracted by overexpression of ROBO1 in HCC cells.
Conclusion: ROBO1 expression is positively correlated with multiple immune checkpoint molecules, suggesting that ROBO1 may be a potential drug target to enhance the potency of immunotherapy. The miR-152-3p/ROBO1 signalling axis contributes to malignant progression and provides a prospective immunotherapeutic target for HCC.
Keywords Roundabout guidance receptor 1, Prognosis, miR-152-3p, Immunotherapy, Hepatic tumorigenesis
Address and Contact Information Department of General Surgery, Afliated Hospital of Chifeng University, No. 42 Wangfu Street, Songshan, Chifeng 024005, China
*Correspondeing author: tao_y717@163.com
Read full article at BMC

No. 23 DOI: 10.1186/s11658-022-00327-7 Volume 27 (2022) - 27:23
Title ROR2 increases the chemoresistance of melanoma by regulating p53 and Bcl2‐family proteins via ERK hyperactivation
Authors María Victoria Castro1,2, Gastón Alexis Barbero1,2, Paula Máscolo1, Rocío Ramos1, María Josefna Quezada1,2 and Pablo Lopez‐Bergami1,2*
Abstract Background: ROR2 is a tyrosine-kinase receptor whose expression is dysregulated in many human diseases. In cancer, ROR2 stimulates proliferation, survival, migration, and metastasis, and is associated with more aggressive tumor stages. The purpose of this work is to study the role of ROR2 in the chemoresistance of melanoma.
Methods: Gain- and loss-of-function experiments were used to study the biological function of ROR2 in melanoma. Cell death induced by chemotherapeutic drugs and BH-3 mimetics was evaluated using crystal violet cytotoxicity assays and annexin V/propidium iodide staining. Western blots were used to evaluate the expression of proteins implicated in cell death. The diferences observed between cells with manipulation of ROR2 levels and control cells were evaluated using both Student’s t-test and ANOVA.
Results: We describe that ROR2 contributes to tumor progression by enhancing the resistance of melanoma cells to both chemotherapeutic drugs and BH-3 mimetics. We demonstrate that ROR2 reduced cell death upon treatment with cisplatin, dacarbazine, lomustine, camptothecin, paclitaxel, ABT-737, TW-37, and venetoclax. This efect was mediated by the inhibition of apoptosis. In addition, we investigated the molecular mechanisms implicated in this role of ROR2. We identifed the MDM2/p53 pathway as a novel target of ROR2 since ROR2 positively regulates MDM2 levels, thus leading to p53 downregulation. We also showed that ROR2 also upregulates Mcl-1 and Bcl2-xL while it negatively regulates Bax and Bid expression. The efect of ROR2 on the expression of these proteins is mediated by the hyperactivation of ERK.
Conclusions: These results demonstrate that ROR2 contributes to melanoma progression by inhibiting apoptosis and increasing chemoresistance. These results not only position ROR2 as a marker of chemoresistance but also support its use as a novel therapeutic target in cancer.
Keywords ROR2, ERK, Melanoma, Chemoresistance, Apoptosis
Address and Contact Information 1 Centro de Estudios Biomédicos, Básicos, Biotecnológicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602, 1405 Buenos Aires, Argentina
2 Consejo Nacional de Investigaciones Científcas y Técnicas (CONICET), 1425 Buenos Aires, Argentina.
*Corresponding author: lopezbergami.pablo@maimonides.edu
Read full article at BMC

No. 24 DOI: 10.1186/s11658-022-00321-z Volume 27 (2022) - 27:24
Title
Authors Junqiang Li1†, Yang Song1†, Chao Zhang1†, Ronglin Wang1, Lei Hua1, Yongdong Guo1, Dongxue Gan1, Liaoliao Zhu1, Shanshan Li1, Peixiang Ma1, Cheng Yang1, Hong Li1, Jing Yang1, Jingjie Shi1, Xiaonan Liu2* and Haichuan Su1*
Abstract Background: Transmembrane protein 43 (TMEM43), a member of the transmembrane protein subfamily, plays a critical role in the initiation and development of cancers. However, little is known concerning the biological function and molecular mechanisms of TMEM43 in pancreatic cancer.
Methods: In this study, TMEM43 expression levels were analyzed in pancreatic cancer samples compared with control samples. The relationship of TMEM43 expression and disease-free survival (DFS) and overall survival (OS) were assessed in pancreatic cancer patients. In vitro and in vivo assays were performed to explore the function and role of TMEM43 in pancreatic cancer. Coimmunoprecipitation (co-IP) followed by protein mass spectrometry was applied to analyze the molecular mechanisms of TMEM43 in pancreatic cancer.
Results: We demonstrated that TMEM43 expression level is elevated in pancreatic cancer samples compared with control group, and is correlated with poor DFS and OS in pancreatic cancer patients. Knockdown of TMEM43 inhibited pancreatic cancer progression in vitro, decreased the percentage of S phase, and inhibited the tumorigenicity of pancreatic cancer in vivo. Moreover, we demonstrated that TMEM43 promoted pancreatic cancer progression by stabilizing PRPF3 and regulating the RAP2B/ERK axis.
Conclusions: The present study suggests that TMEM43 contributes to pancreatic cancer progression through the PRPF3/RAP2B/ERK axis, and might be a novel therapeutic target for pancreatic cancer.
Keywords Pancreatic cancer, Progression, TMEM43, PRPF3, RAP2B
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
*Correspondence: 15353589999@163.com;suhc@fmmu.edu.cn
Junqiang Li, Yang Song and Chao Zhang contributed equally to this work
Read full article at BMC

No. 25 DOI: 10.1186/s11658-022-00326-8 Volume 27 (2022) - 27:25
Title MiR‐5195‐3p FUNCTIONS AS A TUMOR SUPPRESSOR IN PROSTATE CANCER VIA TARGETING CCNL1
Authors Xing Zeng, Zhiquan Hu, Yuanqing Shen, Xian Wei, Jiahua Gan and Zheng Liu*
Abstract Background: Accumulating evidence indicates that miR-5195-3p exerts tumor-suppressive roles in several tumors. However, the clinical signifcance and biological function of miR-5195-3p in prostate cancer (PCa) have not been reported yet.
Methods: The expression levels of miR-5195-3p and Cyclin L1 (CCNL1) were determined using quantitative real-time PCR in clinical specimens and cell lines. The clinical signifcance of miR-5195-3p in patients with PCa was evaluated using Kaplan–Meier survival analysis and Cox regression models. Cell proliferation and cell cycle distribution were measured by CCK-8 assay and fow cytometry, respectively. The association between miR-5195-3p and CCNL1 was analyzed by luciferase reporter assay.
Results: MiR-5195-3p expression levels were signifcantly downregulated in 69 paired PCa tissues compared with matched adjacent normal tissues. The decreased miR-5195-3p expression was associated with Gleason score and TNM stage, as well as worse survival prognosis. The in vitro experiments showed that miR-5195-3p overexpression suppressed the proliferation and cell cycle G1/S transition in PC-3 and DU145 cells. Elevated miR-5195-3p abundance obviously impaired tumor formation in vivo using PC-3 xenografts. Mechanistically, CCNL1 was a direct target of miR-5195-3p in PCa cells, which was inversely correlated with miR-5195-3p in PCa tissues. Importantly, CCNL1 knockdown imitated, while overexpression reversed, the efects of miR-5195-3p overexpression on PCa cell proliferation and cell cycle G1/S transition.
Conclusions: Our data suggest that miR-5195-3p functions as a tumor suppressor by targeting CCNL1 in PCa.
Keywords Prostate cancer, miR-5195-3p, CCNL1, Proliferation
Address and Contact Information Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Ave, Wuhan 430030, Hubei, China
*Corresponding author: liu_zheng0205@126.com
Read full article at BMC

No. 26 DOI: 10.1186/s11658-022-00329-5 Volume 27 (2022) - 27:26
Title ALKBH5 PROMOTES LUNG FBROBLAST ACTIVATION AND SILICA‐INDUCED PULMONARY FBROSIS THROUGH miR‐320a‐3p AND FOXM1
Authors Wenqing Sun1†, Yan Li1†, Dongyu Ma1†, Yi Liu2, Qi Xu1, Demin Cheng1, Guanru Li1 and Chunhui Ni1*
Abstract Background: N6-methyladenosine (m6A) is the most common and abundant internal modifcation of RNA. Its critical functions in multiple physiological and pathological processes have been reported. However, the role of m6A in silica-induced pulmonary fbrosis has not been fully elucidated. AlkB homolog 5 (ALKBH5), a well-known m6A demethylase, is upregulated in the silica-induced mouse pulmonary fbrosis model. Here, we sought to investigate the function of ALKBH5 in pulmonary fbrosis triggered by silica inhalation. Methods: We performed studies with fbroblast cell lines and silica-induced mouse pulmonary fbrosis models. The expression of ALKBH5, miR-320a-3p, and forkhead box protein M1 (FOXM1) was determined by quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RNA immunoprecipitation (RIP) assays and m6 A RNA immuno-precipitation assays (MeRIP), western bolt, immunofuorescence assays, and 5-ethynyl-2’-deoxyuridine (EdU) fuorescence staining were performed to explore the roles of ALKBH5, miR-320a-3p, and FOXM1 in fibroblast activation. Results: ALKBH5 expression was increased in silica-inhaled mouse lung tissues and transforming growth factor (TGF)-β1-stimulated fbroblasts. Moreover, ALKBH5 knockdown exerted antifbrotic efects in vitro. Simultaneously, downregulation of ALKBH5 elevated miR-320a-3p but decreased pri-miR-320a-3p. Mechanically, ALKBH5 demethylated pri-miR-320a-3p, thus blocking the microprocessor protein DGCR8 from interacting with pri-miR-320a-3p and leading to mature process blockage of pri-miR-320a-3p. We further demonstrated that miR-320a-3p could regulate fbrosis by targeting FOXM1 messenger RNA (mRNA) 3′-untranslated region (UTR). Notably, our study also verifed that ALKBH5 could also directly regulate FOXM1 in an m6A-dependent manner. Conclusions: Our fndings suggest that ALKBH5 promotes silica-induced lung fbrosis via the miR-320a-3p/FOXM1 axis or targeting FOXM1 directly. Approaches aimed at ALKBH5 may be efcacious in treating lung fbrosis.
Keywords Silicosis, ALKBH5, miR-320a-3p, FOXM1, m6A
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 Gusu School, Nanjing Medical University, Nanjing 211166, China.
*Corresponding author: chni@njmu.edu.cn; chninjmu@126.com
Wenqing Sun, Yan Li, and Dongyu Ma contributed equally to this work and should be considered co-frst authors
Read full article at BMC

No. 27 DOI: 10.1186/s11658-022-00325-9 Volume 27 (2022) - 27:27
Title INTERLEUKIN‐10 GENETICALLY MODIFED CLINICAL‐GRADE MESENCHYMAL STROMAL CELLS MARKEDLY REINFORCED FUNCTIONAL RECOVERY AFTER SPINAL CORD INJURY VIA DIRECTING ALTERNATIVE ACTIVATION OF MACROPHAGES
Authors Tianyun Gao, Feifei Huang, Wenqing Wang, Yuanyuan Xie and Bin Wang*
Abstract Background: After spinal cord injury (SCI), dysregulated or nonresolving infammatory processes can severely disturb neuronal homeostasis and drive neurodegeneration. Although mesenchymal stromal cell (MSC)-based therapies have showed certain therapeutic efcacy, no MSC therapy has reached its full clinical goal. In this study, we examine interleukin-10 (IL10) genetically modifed clinical-grade MSCs (IL10-MSCs) and evaluate their clinical safety, efectiveness, and therapeutic mechanism in a completely transected SCI mouse model.
Methods: We established stable IL10-overexpressing human umbilical-cord-derived MSCs through electric transduction and screened out clinical-grade IL10-MSCs according to the criteria of cell-based therapeutic products, which were applied to mice with completely transected SCI by repeated tail intravenous injections. Then we comprehensively investigated the motor function, histological structure, and nerve regeneration in SCI mice, and further explored the potential therapeutic mechanism after IL10-MSC treatment.
Results: IL10-MSC treatment markedly reinforced locomotor improvement, accompanied with decreased lesion volume, regeneration of axons, and preservation of neurons, compared with naïve unmodifed MSCs. Further, IL10-MSC transplantation increased the ratio of microglia to infltrated alternatively activated macrophages (M2), and reduced the ratio of classically activated macrophages (M1) at the injured spinal cord, meanwhile increasing the percentage of Treg and Th2 cells, and reducing the percentage of Th1 cells in the peripheral circulatory system. In addition, IL10-MSC administration could prevent apoptosis and promote neuron diferentiation of neural stem cells (NSCs) under infammatory conditions in vitro.
Conclusions: IL10-MSCs exhibited a reliable safety profle and demonstrated promising therapeutic efcacy in SCI compared with naïve MSCs, providing solid support for future clinical application of genetically engineered MSCs.
Keywords MSCs, Interleukin 10, Gene modifcation, Spinal cord injury, Cell-based therapy, Cell quality assessment
Address and Contact Information Center for Clinic Stem Cell Research, the Afliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
*Corresponding author: wangbin022800@126.com
Tianyun Gao and Feifei Huang contributed equally to this study
Read full article at BMC

No. 28 DOI: 10.1186/s11658-022-00324-w Volume 27 (2022) - 27:28
Title PARECOXIB INHIBITS ESOPHAGEAL SQUAMOUS CELL CARCINOMA PROGRESSION VIA THE PDK1–AKT PATHWAY
Authors Han‐Ming Huang1, Xiao‐Yu Huang1, Shao‐Ping Wu1, Can‐Keng Chen1, Xin‐Hua He2* and Yong‐Fa Zhang1*
Abstract Background: Parecoxib plays an important role in inhibition of human cancer. However, the efect of parecoxib on esophageal squamous cell carcinoma (ESCC) is still not well known. The purpose of this study was to investigate the efect of parecoxib on ESCC and its underlying mechanism.
Methods: RNA-sequence analysis was performed to identify functional alterations and mechanisms. Cell cycle, proliferation, invasion, and migration were assessed using fow cytometry, CCK-8 assay, colony formation, transwell, and wound healing assays. Extracellular matrix (ECM) degradation was detected by substrate gel zymography and 3D cell culture assay. Western blotting was used to detect parecoxib-dependent mechanisms involving cell cycle, proliferation, invasion, and migration. Tumor formation in vivo was detected by mouse assay.
Results: Functional experiments indicated that parecoxib induced ESCC cell cycle arrest in G2 phase, and inhibited cell proliferation, invasion, and migration in vitro. Western blotting revealed that parecoxib downregulated the phosphorylation levels of AKT and PDK1, as well as the expression of the mutant p53, cyclin B1, and CDK1, while upregulating p21waf1. Parecoxib inhibited matrix metalloproteinase-2 (MMP2) secretion and invadopodia formation, which were related to ECM degradation. Furthermore, we found that parecoxib suppressed ESCC growth in heterotopic tumor models.
Conclusion: Parecoxib inhibits ESCC progression, including cell cycle, proliferation, invasion, and migration, via the PDK1–AKT signaling pathway.
Keywords Parecoxib, Mutant p53, PDK1–AKT, ESCC
Address and Contact Information 1 Department of Anesthesiology, Second Afliated Hospital of Shantou University Medical College, Shantou 515041, People’s Republic of China
2 Department of Physiology, Shantou University Medical College, Shantou 515041, People’s Republic of China
*Corresponding author: Hexh@stu.edu.cn; 10yfzhang1@stu.edu.cn
Read full article at BMC

No. 29 DOI: 10.1186/s11658-022-00318-8 Volume 27 (2022) - 27:29
Title OBACUNONE ALLEVIATES FERROPTOSIS DURING LIPOPOLYSACCHARIDE‐INDUCED ACUTE LUNG INJURY BY UPREGULATING Nrf2‐DEPENDENT ANTIOXIDANT RESPONSES
Authors Jin Li1,2†, Shi‐hua Deng1,2†, Jing Li1,2†, Li Li1,2, Feng Zhang1,2, Ye Zou1,2, Dong‐ming Wu1,2* and Ying Xu1,2*
Abstract Background: Acute lung injury (ALI) has received considerable attention in the feld of intensive care as it is associated with a high mortality rate. Obacunone (OB), widely found in citrus fruits, is a natural bioactive compound with anti-infammatory and antioxidant activities. However, it is not clear whether OB protects against lipopolysaccharide (LPS)-induced ALI. Therefore, in this study, we aimed to evaluate the protective efects of OB and the potential mechanisms against LPS-induced ALI and BEAS-2B cell injury.
Methods: We established a model of BEAS-2B cell injury and a mouse model of ALI by treating with LPS. Samples of in vitro model were subjected to cell death, Cell Counting Kit-8, and lactate dehydrogenase (LDH) release assays. The total number of cells and neutrophils, protein content, and levels of IL-6, TNF-α, and IL-1β were determined in bronchoalveolar lavage fuid (BALF). Glutathione, reactive oxygen species, and malondialdehyde levels were determined in lung tissue. Additionally, immunohistochemical analysis, immunofuorescence, western blot, quantitative real-time PCR, and enzymelinked immunosorbent assay were conducted to examine the efects of OB. Furthermore, mice were treated with an Nrf2 inhibitor (ML385) to verify its role in ferroptosis. Data were analyzed using one-way analysis of variance or paired t-tests.
Results: Compared with the LPS group, OB efectively alleviated LPS-induced ALI by decreasing lung wet/dry weight ratio, reactive oxygen species and malondialdehyde production, and superoxide dismutase and glutathione consumption in vivo. In addition, OB signifcantly alleviated lung histopathological injury, reduced infammatory cytokine secretion and Fe2+ and 4-HNE levels, and upregulated GPX4, SLC7A11, and Nrf2 expression. Mechanistically, OB activated Nrf2 by inhibiting Nrf2 ubiquitinated proteasome degradation. ML385 reversed the protective efects of OB against LPS-induced ALI.
Conclusion: Overall, OB alleviates LPS-induced ALI, making it a potential novel protective agent against LPS-induced ALI.
Keywords Obacunone, Nrf2, Ferroptosis, Acute lung injury, Lipopolysaccharide
Address and Contact Information 1 The First Afliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, People’s Republic of China.
2 School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan 610500, People’s Republic of China.
*Corresponding author: harvey1989@126.com; yingxu825@126.com
Jin Li, Shi-hua Deng, and Jing Li contributed equally to this work and share first authorship
Read full article at BMC

No. 30 DOI: 10.1186/s11658-022-00328-6 Volume 27 (2022) - 27:30
Title SETD8 COOPERATES WITH MZF1 TO PARTICIPATE IN HYPERGLYCEMIA‐INDUCED ENDOTHELIAL INFAMMATION VIA ELEVATION OF WNT5A LEVELS IN DIABETIC NEPHROPATHY
Authors Fei Wang1†, Wenting Hou1†, Xue Li1, Lihong Lu1, Ting Huang1, Minmin Zhu1,2* and Changhong Miao3*
Abstract Objective: Diabetic nephropathy (DN) is regarded as the main vascular complication of diabetes mellitus, directly afecting the outcome of diabetic patients. Infammatory factors were reported to participate in the progress of DN. Wingless-type family member 5 (WNT5A), myeloid zinc fnger 1 (MZF1), and lysine methyltransferase 8 (SETD8) have also been reported to elevate infammatory factor levels and activate the nuclear factor kappa B (NF-κB) pathway to induce endothelial dysfunction. In the current study, it was assumed that MZF1 associates with SETD8 to regulate WNT5A transcription, thus resulting in hyperglycemia-induced glomerular endothelial infammation in DN.
Methods: The present study recruited 25 diagnosed DN patients (type 2 diabetes) and 25 control participants (nondiabetic renal cancer patients with normal renal function, stage I–II) consecutively. Moreover, a DN rat and cellular model was constructed in the present study. Immunohistochemistry, Western blot, and quantitative polymerase chain reaction (qPCR) were implemented to determine protein and messenger RNA (mRNA) levels. Coimmunoprecipitation (CoIP) and immunofuorescence were implemented in human glomerular endothelial cells (HGECs). Chromatin immunoprecipitation assays and dual luciferase assays were implemented to determine transcriptional activity.
Results: The results of this study indicated that levels of WNT5A expression, p65 phosphorylation (p-p65), and infammatory factors were all elevated in DN patients and rats. In vitro, levels of p-p65 and infammatory factors increased along with the increase of WNT5A expression in hyperglycemic HGECs. Moreover, high glucose increased MZF1 expression and decreased SETD8 expression. MZF1 and SETD8 inhibit each other under the stimulus of high glucose, but cooperate to regulate WNT5A expression, thus infuencing p-p65 and endothelial infammatory factors levels. Overexpression of MZF1 and silencing of SETD8 induced endothelial p-p65 and infammatory factors levels, which can be reversed by si-WNT5A. Mechanistic research indicated that MZF1, SETD8, and its downstream target histone H4 lysine 20 methylation (H4K20me1) all occupied the WNT5A promoter region. sh-SETD8 expanded the enrichment of MZF1 on WNT5A promoter. Our in vivo study proved that SETD8 overexpression inhibited levels of WNT5A, p-p65 expression, and infammatory factors in DN rats.
Conclusions: MZF1 links with SETD8 to regulate WNT5A expression in HGECs, thus elevating levels of hyperglycemia-mediated infammatory factors in glomerular endothelium of DN patients and rats.
Keywords Diabetic nephropathy, SETD8, Endothelial
Address and Contact Information 1 Department of Anesthesiology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
2 Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, People’s Republic of China.
3 Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
*Corresponding author: zhu_mm@126.com; miao_chh@126.com
Fei Wang and Wenting Hou contributed equally to this work
Read full article at BMC

No. 31 DOI: 10.1186/s11658-022-00330-y Volume 27 (2022) - 27:31
Title CircRbms1 KNOCKDOWN ALLEVIATES HYPOXIA‐INDUCED CARDIOMYOCYTE INJURY VIA REGULATING THE miR‐742‐3p/FOXO1 AXIS
Authors Bo Liu and Kai Guo*
Abstract Background: Circular RNA (circRNA) has been shown to play an important role in a variety of cardiovascular diseases, including myocardial infarction (MI). However, the role of circRbms1 in MI progression remains unclear.
Methods: An MI mouse model was constructed in vivo, and cardiomyocytes were cultured under hypoxia condition to induce a cardiomyocyte injury model in vitro. The expression levels of circRbms1, microRNA (miR)-742-3p, and forkhead box O1 (FOXO1) were determined by quantitative real-time PCR. Cell viability, migration, invasion, and apoptosis were measured using Cell Counting Kit-8 assay, transwell assay, and flow cytometry. Meanwhile, western blot analysis was used to examine the protein levels of apoptosis markers and FOXO1. Additionally, dual-luciferase reporter assay, RNA pull-down assay, and RIP assay were employed to verify the interactions between miR-742-3p and circRbms1 or FOXO1.
Results: CircRbms1 was upregulated in the heart tissues of MI mice and hypoxia-induced cardiomyocytes. Hypoxia induced cardiomyocyte injury by suppressing cell viability, migration, and invasion, and promoting apoptosis. Function experiments showed that circRbms1 overexpression aggravated hypoxia-induced cardiomyocyte injury, while its silencing relieved cardiomyocyte injury induced by hypoxia. Furthermore, circRbms1 sponged miR-742-3p. MiR-742-3p overexpression alleviated hypoxia-induced cardiomyocyte injury, and its inhibitor reversed the suppressive efect of circRbms1 silencing on hypoxia-induced cardiomyocyte injury. Further experiments showed that FOXO1 was a target of miR-742-3p, and its expression was positively regulated by circRbms1. The inhibitory efect of miR-742-3p on hypoxia-induced cardiomyocyte injury was reversed by FOXO1 overexpression.
Conclusion: CircRbms1 regulated the miR-742-3p/FOXO1 axis to mediate hypoxia-induced cardiomyocyte injury, suggesting that circRbms1 might be an efective target for MI treatment.
Keywords Myocardial infarction, Hypoxia, CircRbms1, MiR-742-3p, FOXO1
Address and Contact Information Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665 Kongjiang Road, 20092 Shanghai, China
*Corresponding: guokai@xinhuamed.com.cn
Read full article at BMC

No. 32 DOI: 10.1186/s11658-022-00334-8 Volume 27 (2022) - 27:32
Title TIGHT ASSOCIATION OF AUTOPHAGY AND CELL CYCLE IN LEUKEMIA CELLS
Authors Alena Gschwind1,2†, Christian Marx3†, Marie D. Just1,2, Paula Severin1,2, Hannah Behring1,2, Lisa Marx‐Blümel1,2, Sabine Becker1,2, Linda Rothenburger3, Martin Förster4, James F. Beck1 and Jürgen Sonnemann1,2,5*
Abstract Background: Autophagy plays an essential role in maintaining cellular homeostasis and in the response to cellular stress. Autophagy is also involved in cell cycle progression, yet the relationship between these processes is not clearly defned.
Results: In exploring this relationship, we observed that the inhibition of autophagy impaired the G2/M phase-arresting activity of etoposide but enhanced the G1 phase-arresting activity of palbociclib. We further investigated the connection of basal autophagy and cell cycle by utilizing the autophagosome tracer dye Cyto-ID in two ways. First, we established a double-labeling fow-cytometric procedure with Cyto-ID and the DNA probe DRAQ5, permitting the cell cycle phase-specifc determination of autophagy in live cells. This approach demonstrated that diferent cell cycle phases were associated with diferent autophagy levels: G1-phase cells had the lowest level, and G2/M-phase cells had the highest one. Second, we developed a fow-cytometric cell-sorting procedure based on Cyto-ID that separates cell populations into fractions with low, medium, and high autophagy. Cell cycle analysis of Cyto-ID-sorted cells confrmed that the high-autophagy fraction contained a much higher percentage of G2/M-phase cells than the low-autophagy fraction. In addition, Cyto-ID-based cell sorting also proved to be useful for assessing other autophagy-related processes: extracellular fux analysis revealed metabolic diferences between the cell populations, with higher autophagy being associated with higher respiration, higher mitochondrial ATP production, and higher glycolysis.
Conclusion: This work provides clear evidence of high autophagy in G2/M-phase cells by establishing a novel cell sorting technique based on Cyto-ID.
Keywords Autophagy, Cell cycle, Cell sorting, Cyto-ID, DRAQ5, Metabolic analysis
Address and Contact Information 1 Department of Pediatric Hematology and Oncology, Children’s Clinic, Jena University Hospital, Jena, Germany.
2 Research Center Lobeda, Jena University Hospital, 07747 Jena, Germany.
3 Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), 07747 Jena, Germany.
4 Clinic of Internal Medicine I, Jena University Hospital, 07747 Jena, Germany.
5 Klinik für Kinder und Jugendmedizin, Universitätsklinikum Jena, Am Klinikum 1, 07747 Jena, Germany.
*Corresponding author: juergen.sonnemann@med.uni-jena.de
Alena Gschwind and Christian Marx have contributed equally to this work
Read full article at BMC

No.  33DOI: 10.1186/s11658-022-00332-w Volume 27 (2022) - 27:33
Title ANTI‐CANCER PEPTIDE‐BASED THERAPEUTIC STRATEGIES IN SOLID TUMORS
Authors Mohsen Karami Fath1, Kimiya Babakhaniyan2, Maryam Zokaei3,4, Azadeh Yaghoubian5, Sadaf Akbari6, Mahdieh Khorsandi7, Asma Soof8, Mohsen Nabi‐Afadi9, Hamidreza Zalpoor10,11,12, Fateme Jalalifar16, Ali Azargoonjahromi13, Zahra Payandeh14* and Armina Alagheband Bahrami15*
Abstract Background: Nowadays, conventional medical treatments such as surgery, radiotherapy, and chemotherapy cannot cure all types of cancer. A promising approach to treat solid tumors is the use of tumor-targeting peptides to deliver drugs or active agents selectively.
Result: Introducing benefcial therapeutic approaches, such as therapeutic peptides and their varied methods of action against tumor cells, can aid researchers in the discovery of novel peptides for cancer treatment. The biomedical applications of therapeutic peptides are highly interesting. These peptides, owing to their high selectivity, specifcity, small dimensions, high biocompatibility, and easy modifcation, provide good opportunities for targeted drug delivery. In recent years, peptides have shown considerable promise as therapeutics or targeting ligands in cancer research and nanotechnology.
Conclusion: This study reviews a variety of therapeutic peptides and targeting ligands in cancer therapy. Initially, three types of tumor-homing and cell-penetrating peptides (CPPs) are described, and then their applications in breast, glioma, colorectal, and melanoma cancer research are discussed.
Keywords Peptide-based strategies, Tumor-homing peptides, Drug delivery, Targeted delivery
Address and Contact Information 1 Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
2 Department of Medical Surgical Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran, Iran.
3 Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
4 Department of Veterinary Medicine, Beyza Branch, Islamic Azad University, Beyza, Iran.
5 Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran.
6 Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
7 Department of Biotechnology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
8 Department of Physical Chemistry, School of Chemistry, College of Sciences, University of Tehran, Tehran, Iran.
9 Department of Biochemistry, Faculty of biological science, Tarbiat Modares University, Tehran, Iran.
10 American Association of Kidney Patients, Tampa, FL, USA.
11 Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
12 Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientifc Education & Research Network (USERN), Tehran, Iran.
13 Shiraz University of Medical Sciences, Shiraz, Iran.
14 Department Medical Biochemistry and Biophysics, Division Medical Infammation Research, Karolinska Institute, Stockholm, Sweden
15 Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
16 School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran. *Corresponding author: zpayandeh58@yahoo.com; aarminaa@gmail.com
Read full article at BMC

No.  34DOI: 10.1186/s11658-022-00335-7 Volume 27 (2022) - 27:34
Title PROTECTIVE EFECTS OF DEXMEDETOMIDINE IN VITAL ORGAN INJURY: CRUCIAL ROLES OF AUTOPHAGY
Authors Shankun Zhao1†, Weizhou Wu2†, Xuezheng Lin3†, Maolei Shen1, Zhenyu Yang3, Sicong Yu3 and Yu Luo3*
Abstract Vital organ injury is one of the leading causes of global deaths. Accumulating studies have demonstrated that dexmedetomidine (DEX) has an outstanding protective efect on multiple organs for its antiinfammatory and antiapoptotic properties, while the underlying molecular mechanism is not clearly understood. Autophagy, an adaptive catabolic process, has been found to play a crucial role in the organ-protective efects of DEX. Herein, we present a frst attempt to summarize all the evidence on the proposed roles of autophagy in the action of DEX protecting against vital organ injuries via a comprehensive review. We found that most of the relevant studies (17/24, 71%) demonstrated that the modulation of autophagy was inhibited under the treatment of DEX on vital organ injuries (e.g. brain, heart, kidney, and lung), but several studies suggested that the level of autophagy was dramatically increased after administration of DEX. Albeit not fully elucidated, the underlying mechanisms governing the roles of autophagy involve the antiapoptotic properties, inhibiting infammatory response, removing damaged mitochondria, and reducing oxidative stress, which might be facilitated by the interaction with multiple associated genes (i.e., hypoxia inducible factor-1α, p62, caspase-3, heat shock 70 kDa protein, and microRNAs) and signaling cascades (i.e., mammalian target of rapamycin, nuclear factor-kappa B, and c-Jun N-terminal kinases pathway). The authors conclude that DEX hints at a promising strategy in the management of vital organ injuries, while autophagy is crucially involved in the protective efect of DEX.
Keywords Dexmedetomidine (DEX), Autophagy, Organ injury, Protection, Mechanism
Address and Contact Information 1 Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang, China.
2 Department of Urology, Maoming People’s Hospital, Maoming 525000, Guangdong, China.
3 Department of Anesthesia Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
*Corresponding author: luoy@tzzxyy.com
Shankun Zhao, Weizhou Wu, and Xuezheng Lin contributed equally to this work
Read full article at BMC

No.  35DOI: 10.1186/s11658-022-00336-6 Volume 27 (2022) - 27:35
Title CRISPR/Cas9 APPLICATION IN CANCER THERAPY: A PIONEERING GENOME EDITING TOOL
Authors Sadegh Shojaei Baghini1, Zhanna R. Gardanova2, Saeme Azizi Hassan Abadi3, Burhan Abdullah Zaman4, Ahmet İlhan5, Navid Shomali6, Ali Adili7,8, Roozbeh Moghaddar9*† and Amirhossein Fakhre Yaseri10*†
Abstract The progress of genetic engineering in the 1970s brought about a paradigm shift in genome editing technology. The clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9 (CRISPR/Cas9) system is a fexible means to target and modify particular DNA sequences in the genome. Several applications of CRISPR/Cas9 are presently being studied in cancer biology and oncology to provide vigorous site-specifc gene editing to enhance its biological and clinical uses. CRISPR’s flexibility and ease of use have enabled the prompt achievement of almost any preferred alteration with greater efciency and lower cost than preceding modalities. Also, CRISPR/Cas9 technology has recently been applied to improve the safety and efcacy of chimeric antigen receptor (CAR)-T cell therapies and defeat tumor cell resistance to conventional treatments such as chemotherapy and radiotherapy. The current review summarizes the application of CRISPR/Cas9 in cancer therapy. We also discuss the present obstacles and contemplate future possibilities in this context.
Keywords Genome editing, Clustered regularly interspaced short palindromic repeats (CRISPR), CRISPR associated protein 9 (Cas9), Cancer treatment
Address and Contact Information 1 Plant Biotechnology Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
2 Department of Psychotherapy, Pirogov Russian National Research Medical University, 1 Ostrovityanova St., 117997 Moscow, Russia.
3 Department of Nursery and Midwifery, Faculty of Laboratory Science, Islamic Azad University of Chalous, Mazandaran, Iran.
4 Basic Sciences Department, College of Pharmacy, University of Duhok, Kurdistan Region, Iraq.
5 Department of Medical Biochemistry, Faculty of Medicine, Cukurova University, Adana, Turkey.
6 Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran.
7 Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran.
8 Senior Adult Oncology Department, Moftt Cancer Center, University of South Florida, Tampa, USA.
9 Department of Pediatric Hematology and Oncology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
10 Faculty of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran.
*Corresponding author: moghaddarroozbeh@gmail.com; R_moghaddar@yahoo.com; Ahfyaseri@gmail.com
Roozbeh Moghaddar and Amirhossein Fakhre Yaseri contributed equally to this work
Read full article at BMC

No.  36DOI: 10.1186/s11658-022-00337-5 Volume 27 (2022) - 27:36
Title RNF7 INHIBITS APOPTOSIS AND SUNITINIB SENSITIVITY AND PROMOTES GLYCOLYSIS IN RENAL CELL CARCINOMA VIA THE SOCS1/JAK/STAT3 FEEDBACK LOOP
Authors Chengwu Xiao, Wei Zhang, Meimian Hua, Huan Chen, Bin Yang, Ye Wang and Qing Yang*
Abstract Background: RING fnger protein 7 (RNF7) is a highly conserved protein that functions as an E3 ubiquitin ligase. RNF7 overexpression is indicated in multiple human cancers, but its role in renal cell carcinoma (RCC) and the mechanisms underlying how it regulates the initiation and progression of RCC have not been explored.
Methods: Bioinformatics analysis, quantitative reverse-transcription polymerase chain reaction (RT-PCR), and Western blot were conducted to determine the expression of RNF7 in RCC tissues and cell lines. Knockdown and overexpression experiments were performed to examine the efects of RNF7 on cell viability, apoptosis, and glycolysis in vitro and on tumor growth in nude mice in vivo.
Results: The elevated RNF7 expression in tumor tissues of patients with RCC was correlated with poor survival. RNF7 overexpression inhibited apoptosis and promoted glycolysis in vitro and increased tumor growth in vivo by activating the JAK/STAT3 signaling pathway by ubiquitination of SOCS1. Moreover, RNF7 overexpression afected the sensitivity of RCC cells to sunitinib. Finally, STAT3 activation was necessary for transcriptional induction of RNF7.
Conclusion: These results demonstrate that RNF7 inhibited apoptosis, promoted glycolysis, and inhibited sunitinib sensitivity in RCC cells via ubiquitination of SOCS1, thus activating STAT3 signaling. These suggest the potential for targeting the RNF7-SOCS1/JAK/STAT3 pathway for RCC treatment.
Keywords Apoptosis, Clear cell renal cell carcinoma, Glycolysis, JAK/STAT3, RNF7, SOCS1
Address and Contact Information *Corresponding author: yangqchsmmu@163.com Department of Urology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Yangpu, Shanghai 200433, China
Read full article at BMC

No.  37DOI: 10.1186/s11658-022-00338-4 Volume 27 (2022) - 27:37
Title LECTINS AND LECTIBODIES: POTENTIAL PROMISING ANTIVIRAL AGENTS
Authors Mohsen Nabi‐Aadi1†, Morteza Heydari2†, Hamidreza Zalpoor3,4,5, Ibrahim Arman6, Arezoo Sadoughi7, Parisa Sahami8 and Safiyeh Aghazadeh9*
Abstract In nature, lectins are widely dispersed proteins that selectively recognize and bind to carbohydrates and glycoconjugates via reversible bonds at specific binding sites. Many viral diseases have been treated with lectins due to their wide range of structures, specificity for carbohydrates, and ability to bind carbohydrates. Through hemagglutination assays, these proteins can be detected interacting with various carbohydrates on the surface of cells and viral envelopes. This review discusses the most robust lectins and their rationally engineered versions, such as lectibodies, as antiviral proteins. Fusion of lectin and antibody’s crystallizable fragment (Fc) of immunoglobulin G (IgG) produces a molecule called a “lectibody” that can act as a carbohydrate-targeting antibody. Lectibodies can not only bind to the surface glycoproteins via their lectins and neutralize and clear viruses or infected cells by viruses but also perform Fc-mediated antibody efector functions. These functions include complement-dependent cytotoxicity (CDC), antibody-dependent cell-mediated cytotoxicity (ADCC), and antibody-dependent cell-mediated phagocytosis (ADCP). In addition to entering host cells, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein S1 binds to angiotensin-converting enzyme 2 (ACE2) and downregulates it and type I interferons in a way that may lead to lung disease. The SARS-CoV-2 spike protein S1 and human immunodeficiency virus (HIV) envelope are heavily glycosylated, which could make them a major target for developing vaccines, diagnostic tests, and therapeutic drugs. Lectibodies can lead to neutralization and clearance of viruses and cells infected by viruses by binding to glycans located on the envelope surface (e.g., the heavily glycosylated SARS-CoV-2 spike protein).
Keywords Lectins, Lectibody, Carbohydrates, Virus envelope, SARS-CoV-2, HIV, EBV, HCV
Address and Contact Information 1 Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
2 Institute of Bio‐chemistry and Biophysics, University of Tehran, Tehran 13145‐1384, Iran.
3 Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
4 Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
5 American Association of Kidney Patients, Tampa, FL, USA.
6 Department of Molecular Biology and Genetics, Faculty of Sciences and Arts, Zonguldak Bulent Ecevit University, Zonguldak, Turkey.
7 Department of Immunology, International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
8 Medical Biology Research Center, Health Technologies Institute, Kermanshah University of Medical Sciences (KUMS), Kermanshah, Iran.
9 Division of Biochemistry, Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran.
*Corresponding author: s.aghazadeh@urmia.ac.ir
Mohsen Nabi-Aadi and Morteza Heydari contributed equally to this work as first authors
Read full article at BMC

No.  38DOI: 10.1186/s11658-022-00339-3 Volume 27 (2022) - 27:38
Title AN OVERVIEW OF CURRENT DRUGS AND PROPHYLACTIC VACCINES FOR CORONAVIRUS DISEASE 2019 (COVID‐19)
Authors Armina Alagheband Bahrami1†, Ali Azargoonjahromi2†, Samin Sadraei3, Aryan Aarabi3, Zahra Payandeh4* and Masoumeh Rajabibazl5*
Abstract Designing and producing an efective vaccine is the best possible way to reduce the burden and spread of a disease. During the coronavirus disease 2019 (COVID-19) pandemic, many large pharmaceutical and biotechnology companies invested a great deal of time and money in trying to control and combat the disease. In this regard, due to the urgent need, many vaccines are now available earlier than scheduled. Based on their manufacturing technology, the vaccines available for COVID-19 (severe acute respiratory syndrome coronavirus 2 (SAR-CoV2)) infection can be classifed into four platforms: RNA vaccines, adenovirus vector vaccines, subunit (protein-based) vaccines, and inactivated virus vaccines. Moreover, various drugs have been deemed to negatively afect the progression of the infection via various actions. However, adaptive variants of the SARS-CoV-2 genome can alter the pathogenic potential of the virus and increase the difculty of both drug and vaccine development. In this review, along with drugs used in COVID-19 treatment, currently authorized COVID-19 vaccines as well as variants of the virus are described and evaluated, considering all platforms.
Keywords SARS-CoV-2, COVID-19 pandemic, Prophylactic vaccines, Platform, Vaccination
Address and Contact Information 1 Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
2 Shiraz University of Medical Sciences, Shiraz, Iran.
3 Shahid Beheshti University of Medical Sciences, Tehran, Iran.
4 Department Medical Biochemistry and Biophysics, Division Medical Infammation Research, Karolinska Institute, Stockholm, Sweden.
5 Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
*Corresponding author: zahra.payandeh@ki.se; rajabibazl_m@yahoo.com
Armina Alagheband Bahrami and Ali Azargoonjahromi contributed equally to this work.
Read full article at BMC

No.  39DOI: 10.1186/s11658-022-00331-x Volume 27 (2022) - 27:39
Title IDENTIFCATION AND VALIDATION OF AN eight‐lncRNA SIGNATURE THAT PREDICTS PROGNOSIS IN PATIENTS WITH ESOPHAGEAL SQUAMOUS CELL CARCINOMA
Authors Jinfeng Zhang, Xiaodong Ling, Chengyuan Fang and Jianqun Ma*
Abstract Background: Esophageal squamous cell carcinoma (ESCC) is correlated with worse clinical prognosis and lacks available targeted therapy. Thus, identifcation of reliable biomarkers is required for the diagnosis and treatment of ESCC.
Methods: We downloaded the GSE53625 dataset as a training dataset to screen differentially expressed RNAs (DERs) with the criterion of false discovery rate (FDR)<0.05 and |log2fold change (FC)| >1. A support vector machine classifer was used to find the optimal feature gene set that could conclusively distinguish diferent samples. An eight-lncRNA signature was identifed by random survival forest algorithm and multivariate Cox regression analysis. The RNA sequencing data from The Cancer Genome Atlas (TCGA) database were used for external validation. The predictive value of the signature was assessed using Kaplan–Meier test, time-dependent receiver operating characteristic (ROC) curves, and dynamic area under the curve (AUC). Furthermore, a nomogram to predict patients’ 3-year and 5-year prognosis was constructed. CCK-8 assay, fow cytometry, and transwell assay were conducted in ESCC cells.
Results: A total of 1136 DERs, including 689 downregulated mRNAs, 318 upregulated mRNAs, 74 downregulated lncRNAs and 55 upregulated lncRNAs, were obtained in the GES53625 dataset. From the training dataset, we identifed an eight-lncRNA signature, (ADAMTS9-AS1, DLX6-AS1, LINC00470, LINC00520, LINC01497, LINC01749, MAMDC2-AS1, and SSTR5-AS1). A nomogram based on the eight-lncRNA signature, age, and pathologic stage was developed and showed good accuracy for predicting 3-year and 5-year survival probability of patients with ESCC. Functionally, knockdown of LINC00470 signifcantly suppressed cell proliferation, G1/S transition, and migration in two ESCC cell lines (EC9706 and TE-9). Moreover, knockdown of LINC00470 downregulated the protein levels of PCNA, CDK4, and N-cadherin, while upregulating E-cadherin protein level in EC9706 and TE-9 cells.
Conclusion: Our eight-lncRNA signature and nomogram can provide theoretical guidance for further research on the molecular mechanism of ESCC and the screening of molecular markers.
Keywords Esophageal squamous cell carcinoma, Long noncoding RNA, Signature, Nomogram
Address and Contact Information Department of Thoracic Surgery, Esophagus and Mediastinum, Harbin Medical University Cancer Hospital, No.150 Hapin Road, Harbin 150001, Heilongjiang, China
*Corresponding author: jianqun_ma2016@yeah.net
Read full article at BMC

No.  40DOI: 10.1186/s11658-022-00340-w Volume 27 (2022) - 27:40
Title HCMV‐miR‐US33‐5p PROMOTES APOPTOSIS OF AORTIC VASCULAR SMOOTH MUSCLE CELLS BY TARGETING EPAS1/SLC3A2 PATHWAY
Authors Jian Dong1,2*†, Shuangshuang Li2†, Zilin Lu3†, Pengcheng Du2, Guangqin Liu2, Mintao Li1, Chao Ma1, Jian Zhou2* and Junmin Bao2*
Abstract Background: In patients with acute aortic dissection (AAD), increased vascular smooth muscle cell (VSMC) apoptosis has been found. Human cytomegalovirus (HCMV)-miR-US33-5p was signifcantly increased in the plasma of patients with AAD. However, the roles of miR-US33-5p in human aortic VSMC (HA-VSMC) apoptosis remain to be elucidated.
Methods: In the current study, cell apoptosis was analyzed by fow cytometry, cell proliferation by CCK-8 assay, and diferentially expressed genes by RNA sequencing. Luciferase reporter assay was used for binding analysis between miR-US33-5p and endothelial PAS domain protein 1 (EPAS1), and EPAS1 and amino acid transporter heavy chain, member 2 (SLC3A2). The enrichment degree of SLC3A2 promoter DNA was analyzed by chromatin immunoprecipitation assay. Quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and immunoblotting were performed for measuring messenger RNA (mRNA) and protein levels, respectively.
Results: It was found that HCMV infection inhibited proliferation but promoted HA-VSMC apoptosis by upregulating HCMV-miR-US33-5p. Transfection of HCMV-miR-US33-5p mimics the signifcant efect on several signaling pathways including integrin signaling as shown in the RNA sequencing data. Western blotting analysis confrmed that HCMV-miR-US33-5p mimics suppression of the activity of key factors of the integrin signal pathway including FAK, AKT, CAS, and Rac. Mechanistic study showed that HCMV-miR-US33-5p bound to the 3′-untranslated region of EPAS1 to suppress its expression, leading to suppression of SLC3A2 expression, which ultimately promoted cell apoptosis and inhibited cell proliferation. This was confrmed by the fndings that silencing EPAS1 signifcantly reduced the SLC3A2 expression and inhibited proliferation and key factors of integrin signal pathway.
Conclusions: HCMV-miR-US33-5p suppressed proliferation, key factors of integrin signal pathway, and EPAS1/SLC3A2 expression, but promoted HA-VSMC apoptosis. These findings highlighted the importance of HCMV-miR-US33-5p/EPAS1/SCL3A2 signaling and may provide new insights into therapeutic strategies for AAD.
Keywords Acute aortic dissection, Aortic vascular smooth muscle cells, Apoptosis, miRNAs, Endothelial PAS domain protein 1, SLC3A2
Address and Contact Information 1 Department of Vascular Surgery, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
2 Department of Vascular Surgery, Changhai Hospital, Navy Medical University, Shanghai, China.
3 School of Health Science and Engineering, University of Shanghai for Science Technology, Shanghai, China.
*Corresponding author: dr_dongj@163.com; zhoujian1-2@163.com; baojm@189.cn
Jian Dong, Shuangshuang Li, Zilin Lu contributed equally to this work.
Read full article at BMC

No.  41DOI: 10.1186/s11658-022-00342-8 Volume 27 (2022) - 27:41
Title METTL16 PROMOTES HEPATOCELLULAR CARCINOMA PROGRESSION THROUGH DOWNREGULATING RAB11B‐AS1 IN AN m6 A‐DEPENDENT MANNER
Authors Yun‐zhang Dai1, Yong‐da Liu1, Jie Li1, Mei‐ting Chen1, Mei Huang1, Fang Wang1, Qing‐song Yang2*, Ji‐hang Yuan1* and Shu‐han Sun1*
Abstract Background: The molecular mechanisms driving hepatocellular carcinoma (HCC) remain largely unclear. As one of the major epitranscriptomic modifcations, N6-methyladenosine (m6A) plays key roles in HCC. The aim of this study was to investigate the expression, roles, and mechanisms of action of the RNA methyltransferase methyltransferase-like protein 16 (METTL16) in HCC.
Methods: The expression of METTL16 and RAB11B-AS1 was determined by RT-qPCR. The regulation of RAB11B-AS1 by METTL16 was investigated by RNA immunoprecipitation (RIP), methylated RIP (MeRIP), and RNA stability assays. In vitro and in vivo gain- and loss-of-function assays were performed to investigate the roles of METTL16 and RAB11B-AS1.
Results: METTL16 was upregulated in HCC, and its increased expression was correlated with poor prognosis of HCC patients. METTL16 promoted HCC cellular proliferation, migration, and invasion, repressed HCC cellular apoptosis, and promoted HCC tumoral growth in vivo. METTL16 directly bound long noncoding RNA (lncRNA) RAB11B-AS1, induced m6A modifcation of RAB11B-AS1, and decreased the stability of RAB11B-AS1 transcript, leading to the downregulation of RAB11B-AS1. Conversely to METTL16, RAB11B-AS1 is downregulated in HCC, and its decreased expression was correlated with poor prognosis of patients with HCC. Furthermore, the expression of RAB11B-AS1 was negatively correlated with METTL16 in HCC tissues. RAB11B-AS1 repressed HCC cellular proliferation, migration, and invasion, promoted HCC cellular apoptosis, and inhibited HCC tumoral growth in vivo. Functional rescue assays revealed that overexpression of RAB11B-AS1 reversed the oncogenic roles of METTL16 in HCC.
Conclusions: This study identifed the METTL16/RAB11B-AS1 regulatory axis in HCC, which represented novel targets for HCC prognosis and treatment.
Keywords Hepatocellular carcinoma, N6-methyladenosine, RNA methyltransferase, Long noncoding RNA, Tumor progression
Address and Contact Information 1 Department of Medical Genetics, Naval Medical University, Shanghai 200433, China
2 Department of Interventional Radiology, Changhai Hospital, Naval Medical University, Shanghai 20043, China
*Corresponding author: doctoryangqingsong@163.com; jihangyuan@smmu.edu.cn; shsun@vip.sina.com
Read full article at BMC

No.  42DOI: 10.1186/s11658-022-00341-9 Volume 27 (2022) - 27:42
Title COVID‐19 THERAPIES: DO WE SEE SUBSTANTIAL PROGRESS?
Authors Lucyna Matusewicz1†, Marlena Golec2†, Aleksander Czogalla1, Kazimierz Kuliczkowski2, Adam Konka2, Joanna Zembala‐John4,5 and Aleksander F. Sikorski3,5*
Abstract The appearance of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and its spread all over the world is the cause of the coronavirus disease 2019 (COVID-19) pandemic, which has recently resulted in almost 400 million confrmed cases and 6 million deaths, not to mention unknown long-term or persistent side efects in convalescent individuals. In this short review, we discuss approaches to treat COVID-19 that are based on current knowledge of the mechanisms of viral cell receptor recognition, virus–host membrane fusion, and inhibition of viral RNA and viral assembly. Despite enormous progress in antiviral therapy and prevention, new efective therapies are still in great demand.
Keywords ACE2: coronaviruses, COVID-19, COVID-19 therapies, SARS-CoV-2
Address and Contact Information 1 Department of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, ul. F. Joliot Curie 14a, 50‐383 Wrocław, Poland.
2 Silesian Park of Medical Technology Kardio-Med Silesia, ul. M. Curie‐Skłodowskiej 10c, 41‐800 Zabrze, Poland.
3 Research and Development Centre, Regional Specialist Hospital, Kamieńskiego 73a, 51‐154 Wroclaw, Poland.
4 Chair and Department of Medicine and Environmental Epidemiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, H. Jordana 19, 41‐800 Zabrze, Poland.
5 Acellmed Ltd., M. Curie‐Skłodowskiej 10C, 41‐800 Zabrze, Poland.
*Corresponding author: aleksander.sikorski@wssk.wroc.pl
Lucyna Matusewicz and Marlena Golec participated equally in this work.
Read full article at BMC

No.  43DOI: 10.1186/s11658-022-00343-7 Volume 27 (2022) - 27:43
Title MOLECULAR MECHANISM OF LNCRNA SNHG12 IN IMMUNE ESCAPE OF NON‐SMALL CELL LUNG CANCER THROUGH THE HuR/PD‐L1/USP8 AXIS
Authors Yusheng Huang1†, Lei Xia1†, Xiangwu Tan1†, Jingyi Zhang1, Weiwei Zeng1, Benxu Tan1, Xian Yu1, Wei Fang2* and Zhenzhou Yang1*
Abstract Background: The pivotal role of long noncoding RNAs (lncRNAs) in cancer immune responses has been well established. This study was conducted with the aim of exploring the molecular mechanism of lncRNA small nucleolar RNA host gene 12 (SNHG12) in immune escape of non-small cell lung cancer (NSCLC).
Methods: Expression of lncRNA SNHG12, programmed cell death receptor ligand 1 (PD-L1), ubiquitin-specifc protease 8 (USP8), and human antigen R (HuR) in NSCLC tissues and cells was measured, and their binding relationship was determined. NSCLC cell proliferation and apoptosis were assessed. Peripheral blood mononuclear cells (PBMCs) were co-cultured with NSCLC cells. The ratio of CD8+ T cells, PBMC proliferation, and infammatory factors were determined. lncRNA SNHG12 localization was assessed via subcellular fractionation assay. The half-life period of mRNA was determined using actinomycin D. Xenograft tumor models were established to confrm the role of lncRNA SNHG12 in vivo.
Results: LncRNA SNHG12 was found to be prominently expressed in NSCLC tissues and cells, which was associated with a poor prognosis. Silencing lncRNA SNHG12 resulted in the reduction in proliferation and the promotion of apoptosis of NSCLC cells, while simultaneously increasing PBMC proliferation and the ratio of CD8+ T cells. Mechanically, the binding of lncRNA SNHG12 to HuR improved mRNA stability and expression of PD-L1 and USP8, and USP8-mediated deubiquitination stabilized the protein level of PD-L1. Overexpression of USP8 or PD-L1 weakened the inhibition of silencing lncRNA SNHG12 on the immune escape of NSCLC. Silencing lncRNA SNHG12 restricted tumor growth and upregulated the ratio of CD8+ T cells by decreasing USP8 and PD-L1.
Conclusion: LncRNA SNHG12 facilitated the immune escape of NSCLC by binding to HuR and increasing PD-L1 and USP8 levels.
Keywords Non-small cell lung cancer, LncRNA SNHG12, PD-L1, USP8, Immune escape, HuR, Peripheral blood mononuclear cells, CD8+ T
Address and Contact Information 1 Department of Cancer Center, The Second Afliated Hospital of Chongqing Medical University, Tianwen Avenue No. 288, Nan’an District, Chongqing 400010, China
2 Chongqing University, Three Gorges Hospital, No. 165 Xincheng Road, Wanzhou District, Chongqing 404100, China
*Correspondence: lixizi19620716@163.com; yangzhenzhou@sohu.com
Yusheng Huang, Lei Xia, and Xiangwu Tan contributed equally to this work.
Read full article at BMC

No.  44DOI: 10.1186/s11658-022-00345-5 Volume 27 (2022) - 27:44
Title EFECT OF CHRONIC INTERMITTENT HYPOXIA‐INDUCED HIF‐1α/ATAD2 EXPRESSION ON LUNG CANCER STEMNESS
Authors Shengyu Hao1†, Fan Li2†, Pan Jiang2* and Jian Gao2*
Abstract Background: Obstructive sleep apnea is associated with increased lung cancer incidence and mortality. Cancer stem cells (CSCs) are characterized by their self-renewing ability, which contributes to metastasis, recurrence, and drug resistance. ATPase family AAA domain-containing protein 2 (ATAD2) induces malignancy in diferent types of tumors. However, a correlation between ATAD2 expression and CSCs in lung cancer has not yet been reported.
Methods: The relative messenger RNA (mRNA) levels of ATAD2, CD44, CD133, and hypoxia-inducible factor (HIF)-1α were determined using reverse-transcription quantitative polymerase chain reaction. ATAD2 protein levels were determined using Western blotting. Cell counting kit-8, 5-ethynyl-2′-deoxyuridine (EdU), and colony formation assays were performed to analyze the proliferation of lung cancer cells. Transwell migration and invasion assays were performed to evaluate cell migration and invasion, respectively. Tumor sphere formation analysis was used to determine tumor spheroid capacity. The link between ATAD2 and HIF-1α was verifed using a dual-luciferase reporter assay. Immunofuorescence staining was performed to assess mitochondrial reactive oxygen species (mtROS) production. Flow cytometry analysis was conducted to determine the CD133 and CD44 positive cell ratio.
Results: We evaluated the relative expression of ATAD2 in four lung cancer cell lines (A549, SPC-A1, H460, and H1299 cells) and found increased mRNA and protein levels of ATAD2 in lung cancer samples. ATAD2 overexpression was a poor prognostic factor for lung cancer patients. Loss of ATAD2 reduced lung cancer cell viability and proliferation. Additionally, ATAD2 knockdown repressed lung cancer cell migration, invasion, stem-cell-like properties, and mtROS production. Chronic intermittent hypoxia (CIH)-induced HIF-1α expression signifcantly activated ATAD2 during lung cancer progression.
Conclusions: This study found that CIH induced HIF-1α expression, which acts as a transcriptional activator of ATAD2. The present study also suggests a novel mechanism by which the integrity of CIH-triggered HIF-1α/ATAD2 may determine lung cancer aggressiveness via the interplay of mtROS and stemness in lung cancer cells.
Keywords ATAD2, Lung cancer stem cells, CIH, HIF-1α
Address and Contact Information 1 Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
2 Department of Nutrition, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032, China
*Correspondence: jiang.pan@zs-hospital.sh.cn; gao.jian@zs-hospital.sh.cn
Shengyu Hao and Fan Li contributed equally to this work.
Read full article at BMC

No.  45DOI: 10.1186/s11658-022-00351-7 Volume 27 (2022) - 27:45
Title ALPHA‐SMOOTH MUSCLE ACTIN‐POSITIVE CANCER‐ASSOCIATED FBROBLASTS SECRETING OSTEOPONTIN PROMOTE GROWTH OF LUMINAL BREAST CANCER
Authors Anna Muchlińska1, Anna Nagel1, Marta Popęda1, Jolanta Szade2, Magdalena Niemira3, Jacek Zieliński4, Jarosław Skokowski4,5, Natalia Bednarz‐Knoll1 and Anna J. Żaczek1*
Abstract Background: Cancer-associated fbroblasts (CAFs) have been shown to support tumor development in a variety of cancers. Diferent markers were applied to classify CAFs in order to elucidate their impact on tumor progression. However, the exact mechanism by which CAFs enhance cancer development and metastasis is yet unknown.
Methods: Alpha-smooth muscle actin (α-SMA) was examined immunohistochemically in intratumoral CAFs of nonmetastatic breast cancers and correlated with clinico‐pathological data. Four CAF cell lines were isolated from patients with luminal breast cancer (lumBC) and classifed according to the presence of α-SMA protein. Conditioned medium (CM) from CAF cultures was used to assess the infuence of CAFs on lumBC cell lines: MCF7 and T47D cells using Matrigel 3D culture assay. To identify potential factors accounting for promotion of tumor growth by α-SMAhigh CAFs, nCounter PanCancer Immune Profling Panel (NanoString) was used.
Results: In luminal breast cancer, presence of intratumoral CAFs expressing high level of α-SMA (13% of lumBC group) correlated with poor prognosis (p=0.019). In in vitro conditions, conditioned medium obtained from primary cultures of α-SMA-positive CAFs isolated from luminal tumors was observed to enhance growth of lumBC cell line colonies in 3D Matrigel, in contrast to CM derived from α-SMA-negative CAFs. Multigene expression analysis indicated that osteopontin (OPN) was overexpressed in α-SMA-positive CAFs in both clinical samples and in vitro models. OPN expression was associated with higher percentage of Ki67-positive cells in clinical material (p=0.012), while OPN blocking in α-SMA-positive CAF-derived CM attenuated growth of lumBC cell line colonies in 3D Matrigel.
Conclusions: Our fndings demonstrate that α-SMA-positive CAFs might enhance tumor growth via secretion of OPN.
Keywords Cancer-associated fbroblast, Luminal breast cancer, Alpha-smooth muscle actin, Osteopontin
Address and Contact Information 1 Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, 80‐211 Gdansk, Poland
2 Department of Pathomorphology, Medical University of Gdansk, 80‐214 Gdansk, Poland
3 Clinical Research Centre, Medical University of Bialystok, 15‐276 Bialystok, Poland
4 Department of Surgical Oncology, Medical University of Gdansk, 80‐214 Gdansk, Poland
5 Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), 80‐211 Gdansk, Poland
*Correspondence: azaczek@gumed.edu.pl
Read full article at BMC

No.  46DOI: 10.1186/s11658-022-00333-9 Volume 27 (2022) - 27:46
Title REVERSELY IMMORTALIZED MOUSE SALIVARY GLAND CELLS PRESENTED A PROMISING METABOLIC AND FBROTIC RESPONSE UPON BMP9/Gdf2 STIMULATION
Authors Wenping Luo1,2†, Panpan Liang1,3†, Tianyu Zhao3,4, Qianyu Cheng3, Huikai Liu3, Liwen He1,2, Linghuan Zhang5, Bo Huang6, Yuxin Zhang3, Tongchuan He2 and Deqin Yang1,3*
Abstract The submandibular gland (SMG) and the sublingual gland (SLG) are two of the three major salivary glands in mammals. In mice, they are adjacent to each other and open into the oral cavity, producing saliva to lubricate the mouth and aid in food digestion. Though salivary gland dysfunction accompanied with fbrosis and metabolic disturbance is common in clinic, in-depth mechanistic research is lacking. Currently, research on how to rescue salivary function is challenging, as it must resort to using terminally diferentiated acinar cells or precursor acinar cells with unknown diferentiation. In this study, we established reversely immortalized mouse primary SMG cells (iSMGCs) and SLG cells (iSLGCs) on the frst postnatal day (P0). The iSMGCs and iSLGCs grew well, exhibited many salivary gland characteristics, and retained the metabolism-related genes derived from the original tissue as demonstrated using transcriptome sequencing (RNA-seq) analysis. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of these two cell lines, which overlapped with those of the SMG and SLG, were enriched in cysteine and methionine metabolism. Furthermore, we investigated the role of bone morphogenetic protein 9 (BMP9), also known as growth diferentiation factor 2(Gdf2), on metabolic and fbrotic functions in the SMG and SLG. We demonstrated that iSMGCs and iSLGCs presented promising adipogenic and fbrotic responses upon BMP9/Gdf2 stimulation. Thus, our fndings indicate that iSMGCs and iSLGCs faithfully reproduce characteristics of SMG and SLG cells and present a promising prospect for use in future study of salivary gland metabolism and fbrosis upon BMP9/Gdf2 stimulation.
Keywords Submandibular gland, Sublingual gland, Immortalization, Metabolism, Fibrosis, BMP9/Gdf2
Address and Contact Information 1 Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, 426 Songshi North Road, Yubei District, Chongqing 401147, China.
2 Department of Surgery, Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, The University of Chicago Medical Center, 5841 South Maryland Avenue MC6035, Chicago, IL 60637, USA.
3 Stomatological Hospital of Chongqing Medical University, 426 Songshi North Road, Yubei District, Chongqing 401147, China.
4 Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, 426 Songshi North Road, Yubei District, Chongqing 401147, China.
5 Stem Cell Biology and Therapy Laboratory, Ministry of Education Key Laboratory of Child Development and Disorders, The Children’s Hospital of Chongqing Medical University, Chongqing 400014, China.
6 Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, The Second Afliated Hospital of Nanchang University, No.1 Min De Road, Nanchang 330006, China.
*Corresponding author: yangdeqin@hospital.cqmu.edu.cn
Wenping Luo and Panpan Liang equally contributed to this work.
Read full article at BMC

No.  47DOI: 10.1186/s11658-022-00346-4 Volume 27 (2022) - 27:47
Title INTEGRATED ANALYSIS OF tRNA‐DERIVED SMALL RNAs IN PROLIFERATIVE HUMAN AORTIC SMOOTH MUSCLE CELLS
Authors Jian‐Zhi Zhao1,2†, Qi‐Yao Li3†, Jia‐Jie Lin1†, Li‐Yun Yang1, Mei‐Yang Du1, Yu Wang1, Ke‐Xin Liu1, Ze‐An Jiang1, Huan‐Huan Li1, Si‐Fan Wang1, Bo Sun1, Shi‐Qing Mu1, Bin Li1, Kun Liu1, Miao Gong1 and Shao‐Guang Sun1*
Abstract Background: Abnormal proliferation of vascular smooth muscle cells (VSMCs) contributes to vascular remodeling diseases. Recently, it has been discovered that tRNA-derived small RNAs (tsRNAs), a new type of noncoding RNAs, are related to the proliferation and migration of VSMCs. tsRNAs regulate target gene expression through miRNA-like functions. This study aims to explore the potential of tsRNAs in human aortic smooth muscle cell (HASMC) proliferation.
Methods: High-throughput sequencing was performed to analyze the tsRNA expression profle of proliferative and quiescent HASMCs. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to validate the sequence results and subcellular distribution of AS-tDR-001370, AS-tDR-000067, AS-tDR-009512, and AS-tDR-000076. Based on the microRNA-like functions of tsRNAs, we predicted target promoters and mRNAs and constructed tsRNA–promoter and tsRNA–mRNA interaction networks. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to reveal the function of target genes. EdU incorporation assay, Western blot, and dual-luciferase reporter gene assay were utilized to detect the efects of tsRNAs on HASMC proliferation.
Results: Compared with quiescent HASMCs, there were 1838 diferentially expressed tsRNAs in proliferative HASMCs, including 887 with increased expression (fold change>2, p<0.05) and 951 with decreased expression (fold change<1⁄2, p<0.05). AS-tDR-001370, AS-tDR-000067, AS-tDR-009512, and AS-tDR-000076 were increased in proliferative HASMCs and were mainly located in the nucleus. Bioinformatics analysis suggested that the four tsRNAs involved a variety of GO terms and pathways related to VSMC proliferation. AS-tDR-000067 promoted HASMC proliferation by suppressing p53 transcription in a promoter-targeted manner. AS-tDR-000076 accelerated HASMC proliferation by attenuating mitofusin 2 (MFN2) levels in a 3′-untranslated region (UTR)-targeted manner.
Conclusions: During HASMC proliferation, the expression levels of many tsRNAs are altered. AS-tDR-000067 and AS-tDR-000076 act as new factors promoting VSMC proliferation.
Keywords tsRNA, VSMC, proliferation, p53, MFN2
Address and Contact Information 1 Department of Biochemistry and Molecular Biology, Key Laboratory of Medical Biotechnology of Hebei Province, Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, China
2 Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
3Department of Emergency Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
*Corresponding author: sunshaoguang00@163.com
Jian-Zhi Zhao, Qi-Yao Li, and Jia-Jie Lin contributed equally to this work.
Read full article at BMC

No.  48DOI: 10.1186/s11658-022-00353-5 Volume 27 (2022) - 27:48
Title METFORMIN INHIBITS MELANOMA CELL METASTASIS BY SUPPRESSING THE miR‐5100/SPINK5/STAT3 AXIS
Authors Dong Suwei1,2†, Xiao Yanbin2,4*†, Wang Jianqiang1†, Ma Xiang2, Peng Zhuohui2, Kang Jianping2, Wang Yunqing1,4* and Li Zhen3,4*
Abstract Melanoma is the most lethal skin cancer characterized by its high metastatic potential. It is urgent to fnd novel therapy strategies to overcome this feature. Metformin has been confrmed to suppress invasion and migration of various types of cancer. However, additional mechanisms underlying the antimetastatic efect of metformin on melanoma require further investigation. Here, we performed microarray analysis and uncovered an altered mRNA and miRNA expression profle between melanoma and nevus. Luciferase reporter assay confrmed that miR-5100 targets SPINK5 to activate STAT3 phosphorylation. Migration and wound healing assays showed that the miR-5100/SPINK5/STAT3 axis promotes melanoma cell metastasis; the mechanism was proven by initiation of epithelial–mesenchymal transition. Co-immunoprecipitation (Co-IP) further confrmed an indirect interaction between SPINK5 and STAT3. Furthermore, metformin dramatically inhibited miR-5100/SPINK5/STAT3 pathway, and decreased B16-F10 cell metastasis to lung in C57 mouse module. Intriguingly, pretreatment of metformin before melanoma cell injection improved this efect further. These fndings exposed the underlying mechanisms of action of metformin and update the use of this drug to prevent metastasis in melanoma.
Keywords EMT, miR-5100, Metformin, SPINK5, STAT3
Address and Contact Information 1 Department of Orthopaedics, The Second Afliated Hospital of Xuzhou Medical University, Xuzhou 221000, People’s Republic of China
2 Department of Orthopaedics, The Third Afliated Hospital of Kunming Medical University, Kunming 650118, People’s Republic of China
3 Department of Medical Oncology, The Second Afliated Hospital of Xuzhou Medical University, Xuzhou 221000, People’s Republic of China
4 The Second Afliated Hospital of Xuzhou Medical University, Xuzhou 221000, People’s Republic of China
*Corresponding author: xiaoyanbin73@126.com; kzwangyunqing@163.com; lizhenres@163.com
Dong Suwei, Xiao Yanbin and Wang Jianqiang contribute equally to this work
Read full article at BMC

No.  49DOI: 10.1186/s11658-022-00348-2 Volume 27 (2022) - 27:49
Title CRISPR/Cas9 GENE EDITING: A NEW APPROACH FOR OVERCOMING DRUG RESISTANCE IN CANCER
Authors Mostafa Vaghari‐Tabari1, Parisa Hassanpour1, Fatemeh Sadeghsoltani1, Faezeh Malakoti1, Forough Alemi1, Durdi Qujeq2,3, Zatollah Asemi4* and Bahman Yousef1,5*
Abstract The CRISPR/Cas9 system is an RNA-based adaptive immune system in bacteria and archaea. Various studies have shown that it is possible to target a wide range of human genes and treat some human diseases, including cancers, by the CRISPR/Cas9 system. In fact, CRISPR/Cas9 gene editing is one of the most efcient genome manipulation techniques. Studies have shown that CRISPR/Cas9 technology, in addition to having the potential to be used as a new therapeutic approach in the treatment of cancers, can also be used to enhance the efectiveness of existing treatments. Undoubtedly, the issue of drug resistance is one of the main obstacles in the treatment of cancers. Cancer cells resist anticancer drugs by a variety of mechanisms, such as enhancing anticancer drugs efux, enhancing DNA repair, enhancing stemness, and attenuating apoptosis. Mutations in some proteins of diferent cellular signaling pathways are associated with these events and drug resistance. Recent studies have shown that the CRISPR/Cas9 technique can be used to target important genes involved in these mechanisms, thereby increasing the efectiveness of anticancer drugs. In this review article, studies related to the applications of this technique in overcoming drug resistance in cancer cells will be reviewed. In addition, we will give a brief overview of the limitations of the CRISP/Cas9 gene-editing technique.
Keywords CRISPR/Cas9, Gene editing, Chemoresistance, Malignancy, Cancer treatment
Address and Contact Information 1 Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
2 Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran
3 Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
4 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
5 Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
*Correspondence: asemi_r@yahoo.com; yousefb@tbzmed.ac.ir
Read full article at BMC

No.  50DOI: 10.1186/s11658-022-00352-6 Volume 27 (2022) - 27:50
Title SARS‐CoV‐2: PHENOTYPE, GENOTYPE, AND CHARACTERIZATION OF DIFERENT VARIANTS
Authors Mohammadreza Saberiyan1, Elham Karimi2, Zahra Khademi3, Parvaneh Movahhed4, Amir Saf5 and Ameneh Mehri‐Ghahfarrokhi6*
Abstract Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of corona‐virus disease 2019 (COVID-19), a major international public health concern. Because of very similar amino acid sequences of the seven domain names, SARS-CoV-2 belongs to the Coronavirinae subfamily of the family Coronaviridae, order Nidovirales, and realm Riboviria, placed in exceptional clusters, but categorized as a SARS-like species. As the RNA virus family with the longest genome, the Coronaviridae genome consists of a single strand of positive RNA (25–32 kb in length). Four major structural proteins of this genome include the spike (S), membrane (M), envelope (E), and the nucleocapsid (N) protein, all of which are encoded within the 3′ end of the genome. By engaging with its receptor, angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 infects host cells. According to the most recent epidemiological data, as the illness spread globally, several genetic variations of SARS-CoV-2 appeared quickly, with the World Health Organization (WHO) naming 11 of them. Among these, seven SARS-CoV-2 subtypes have received the most attention. Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.617.2) are now designated as variations of concern (VOC) (B.1.1.529). Lambda (C.37) and Mu are variations of interest (VOI) (B.1.621). The remaining six are either being monitored or are no longer considered a threat. On the basis of studies done so far, antiviral drugs, antibiotics, glucocorticoids, recombinant intravenous immunoglobulin, plasma therapy, and IFN-α2b have been used to treat patients. Moreover, full vaccination is associated with lower infection and helps prevent transmission, but the risk of infection cannot be eliminated completely in vaccinated people.
Keywords SARS-CoV-2 variants, Omicron, COVID-19, Vaccine
Address and Contact Information 1 Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
2 Department of Medical Genetics, School of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
3 Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
4 Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
5 Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
6 Clinical Research Development Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
*Corresponding author: ameneh.mehri.96@gmail.com
Read full article at BMC

No.  51DOI: 10.1186/s11658-022-00350-8 Volume 27 (2022) - 27:51
Title WTAP‐MEDIATED N6‐METHYLADENOSINE MODIFCATION OF NLRP3 mRNA IN KIDNEY INJURY OF DIABETIC NEPHROPATHY
Authors Jianzi Lan*, Bowen Xu, Xin Shi, Qi Pan and Qing Tao
Abstract Background: Diabetic nephropathy (DN) is prevalent in patients with diabetes. N6-methyladenosine (m6A) methylation has been found to cause modifcation of nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing (NLRP) 3, which is involved in cell pyroptosis and infammation. WTAP is a key gene in modulating NLRP3 m6A.
Methods: In this study, WTAP was silenced or overexpressed in high glucose (HG)-treated HK-2 cells to determine its infuence on pyroptosis, NLRP3 infammasome-related proteins, and the release of pro-infammatory cytokines. NLRP3 expression and m6A levels were assessed in the presence of WTAP shRNA (shWTAP). WTAP expression in HK-2 cells was examined with the introduction of C646, a histone acetyltransferase p300 inhibitor.
Results: We found that WTAP expression was enhanced in patients with DN and in HG-treated HK-2 cells. Knockdown of WTAP attenuated HG-induced cell pyroptosis and NLRP3-related pro-infammatory cytokines in both HK-2 cells and db/db mice, whereas WTAP overexpression promoted these cellular processes in HK-2 cells. WTAP mediated the m6A of NLRP3 mRNA that was stabilized by insulin-like growth factor 2 mRNA binding protein 1. Histone acetyltransferase p300 regulated WTAP expression. WTAP mRNA levels were positively correlated with NLRP3 infammasome components and pro-infammatory cytokines.
Conclusion: Taken together, WTAP promotes the m6A methylation of NLRP3 mRNA to upregulate NLRP3 infammasome activation, which further induces cell pyroptosis and infammation.
Keywords WTAP, NLRP3, Pyroptosis, High glucose, N6-methyladenosine, Infammation
Address and Contact Information Department of Traditional Chinese Medicine, Shanghai East Hospital, Tongji University School of Medicine, No. 150, Jimo Road, Pudong District, Shanghai 200120, China
*Correspondence: dfsl112@126.com
Read full article at BMC

No.  52DOI:10.1186/s11658-022-00344-6 Volume 27 (2022) - 27:52
Title THE ROLE OF EPIGENETIC MODIFCATIONS IN DRUG RESISTANCE AND TREATMENT OF BREAST CANCER
Authors Mohsen Karami Fath1, Ali Azargoonjahromi2, Arash Kiani3, Fateme Jalalifar4, Parisa Osati5, Mahsa Akbari Oryani6, Fateh Shakeri1, Farhad Nasirzadeh1, Behman Khalesi7, Mohsen Nabi‐Aadi8, Hamidreza Zalpoor9,10, Maysam Mard‐Soltani11* and Zahra Payandeh12*
Abstract Background: Breast cancer is defined as a biological and molecular heterogeneous disorder that originates from breast cells. Genetic predisposition is the most important factor giving rise to this malignancy. The most notable mutations in breast cancer occur in the BRCA1 and BRCA2 genes. Owing to disease heterogeneity, lack of therapeutic target, anti-cancer drug resistance, residual disease, and recurrence, researchers are faced with challenges in developing strategies to treat patients with breast cancer.
Results: It has recently been reported that epigenetic processes such as DNA methylation and histone modification, as well as microRNAs (miRNAs), have potently contributed to the pathophysiology, diagnosis, and treatment of breast cancer. These observa‐tions have persuaded researchers to move their therapeutic approaches beyond the genetic framework toward the epigenetic concept.
Conclusion: Herein we discuss the molecular and epigenetic mechanisms underlying breast cancer progression and resistance as well as various aspects of epigenetic-based therapies as monotherapy and combined with immunotherapy.
Keywords Breast cancer, Epigenetic modifications, microRNAs, Treatment, Chemoresistance
Address and Contact Information 1 Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran.
2 Shiraz University of Medical Sciences, Shiraz, Iran.
3 Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran.
4 School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran.
5 Chemical Engineering Department, Fouman Faculty of Engineering, College of Engineering, University of Tehran, Fouman, Iran.
6 Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
7 Department of Research and Production of Poultry Viral Vaccine, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization, Karaj, Iran.
8 Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran.
9 Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
10 Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
11 Department of Clinical Biochemistry, Faculty of Medical Sciences, Dezful University of Medical Sciences, Dezful, Iran.
12 Department Medical Biochemistry and Biophysics, Division Medical Infammation Research, Karolinska Institute, Stockholm, Sweden.
*Corresponding author: maysam.mardsoltani@gmail.com; Zpayandeh58@yahoo.com
Read full article at BMC

No.  53DOI: 10.1186/s11658-022-00347-3 Volume 27 (2022) - 27:53
Title RAMAN MICROSPECTROSCOPY FNGERPRINTING OF ORGANOID DIFERENTIATION STATE
Authors Kate Tubbesing1,2,3,6, Nicholas Moskwa2,3,5, Ting Chean Khoo1, Deirdre A. Nelson2,3, Anna Sharikova1, Yunlong Feng4, Melinda Larsen2,3* and Alexander Khmaladze1*
Abstract Background: Organoids, which are organs grown in a dish from stem or progenitor cells, model the structure and function of organs and can be used to defne molecular events during organ formation, model human disease, assess drug responses, and perform grafting in vivo for regenerative medicine approaches. For therapeutic applications, there is a need for nondestructive methods to identify the diferentiation state of unlabeled organoids in response to treatment with growth factors or pharmacologicals.
Methods: Using complex 3D submandibular salivary gland organoids developed from embryonic progenitor cells, which respond to EGF by proliferating and FGF2 by undergoing branching morphogenesis and proacinar diferentiation, we developed Raman confocal microspectroscopy methods to defne Raman signatures for each of these organoid states using both fxed and live organoids.
Results: Three separate quantitative comparisons, Raman spectral features, multivariate analysis, and machine learning, classifed distinct organoid diferentiation signatures and revealed that the Raman spectral signatures were predictive of organoid phenotype. Conclusions: As the organoids were unlabeled, intact, and hydrated at the time of imaging, Raman spectral fngerprints can be used to noninvasively distinguish between diferent organoid phenotypes for future applications in disease modeling, drug screening, and regenerative medicine.
Keywords Raman spectroscopy, Tissue-engineered organoids, Salivary gland organoids, Regenerative medicine
Address and Contact Information 1 Department of Physics, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
2 Department of Biological Sciences, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
3 RNA Institute, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
4 Department of Mathematics, University at Albany, State University of New York, 1400 Washington Avenue, Albany, NY 12222, USA
5 Present Address: The Jackson Laboratory, 10 Discovery Dr., Farmington, CT 06032, USA
6 Present Address: Neural Stem Cell Institute, Rensselaer, NY 12144, USA
*Corresponding author: mlarsen@albany.edu; akhmaladze@albany.edu
Read full article at BMC

No.  54DOI: 10.1186/s11658-022-00357-1 Volume 27 (2022) - 27:54
Title CORRECTION TO: ROR2 INCREASES THE CHEMORESISTANCE OF MELANOMA BY REGULATING p53 AND Bcl2‐FAMILY PROTEINS VIA ERK HYPERACTIVATION
Authors María Victoria Castro1,2, Gastón Alexis Barbero1,2, Paula Máscolo1, Rocío Ramos1, María Josefna Quezada1,2 and Pablo Lopez‐Bergami1,2*
Abstract Correction to: Cellular & Molecular Biology Letters (2022) 27:23
https://doi.org/10.1186/s11658-022-00327-7

Following publication of the original article [1], the authors identifed a few errors in panel A of Fig. 4. Two western blot images from panel B (Bcl-xL and Actin) were duplicated by mistake into panel A in place of the western blots for MDM2 and the Actin controls for both MDM2 and p53. Te correct Fig. 4 is given in this correction article.
Keywords ROR2, ERK, Melanoma, Chemoresistance, Apoptosis
Address and Contact Information 1 Centro de Estudios Biomédicos, Básicos, Biotecnológicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602, 1405 Buenos Aires, Argentina
2 Consejo Nacional de Investigaciones Científcas y TécniCas (CONICET), 1425 Buenos Aires, Argentina
*Corresponding author: lopezbergami.pablo@maimonides.edu
The original article can be found online at https://doi.org/10.1186/s11658-​022-​00327-7
Read full article at BMC

No.  55DOI: 10.1186/s11658-022-00349-1 Volume 27 (2022) - 27:55
Title METTL3 MEDIATES Ang‐II‐INDUCED CARDIAC HYPERTROPHY THROUGH ACCELERATING pri‐miR‐221/222 MATURATION IN AN m6A‐DEPENDENT MANNER
Authors Rui Zhang, Yangyang Qu, Zhenjun Ji, Chunshu Hao, Yamin Su, Yuyu Yao, Wenjie Zuo, Xi Chen, Mingming Yang and Genshan Ma*
Abstract Background: METTL3 is the core catalytic enzyme in m6A and is involved in a variety of cardiovascular diseases. However, whether and how METTL3 plays a role during angiotensin II (Ang-II)-induced myocardial hypertrophy is still unknown.
Methods: Neonatal rat cardiomyocytes (NRCMs) and C57BL/6J mice were treated with Ang-II to induce myocardial hypertrophy. qRT-PCR and western blots were used to detect the expression of RNAs and proteins. Gene function was verifed by knockdown and/or overexpression, respectively. Luciferase and RNA immunoprecipitation (RIP) assays were used to verify interactions among multiple genes. Wheat germ agglutinin (WGA), hematoxylin and eosin (H&E), and immunofuorescence were used to examine myocardial size. m6A methylation was detected by a colorimetric kit.
Results: METTL3 and miR-221/222 expression and m6A levels were signifcantly increased in response to Ang-II stimulation. Knockdown of METTL3 or miR-221/222 could completely abolish the ability of NRCMs to undergo hypertrophy. The expression of miR-221/222 was positively regulated by METTL3, and the levels of pri-miR-221/222 that bind to DGCR8 or form m6A methylation were promoted by METTL3 in NRCMs. The efect of METTL3 knockdown on hypertrophy was antagonized by miR-221/222 overexpression. Mechanically, Wnt/β-catenin signaling was activated during hypertrophy and restrained by METTL3 or miR-221/222 inhibition. The Wnt/β-catenin antagonist DKK2 was directly targeted by miR-221/222, and the efect of miR-221/222 inhibitor on Wnt/β-catenin was abolished after inhibition of DKK2. Finally, AAV9-mediated cardiac METTL3 knockdown was able to attenuate Ang-II-induced cardiac hypertrophy in mouse model.
Conclusions: Our fndings suggest that METTL3 positively modulates the pri-miR221/222 maturation process in an m6A-dependent manner and subsequently activates Wnt/β-catenin signaling by inhibiting DKK2, thus promoting Ang-II-induced cardiac hypertrophy. AAV9-mediated cardiac METTL3 knockdown could be a therapeutic for pathological myocardial hypertrophy.
Keywords Cardiac hypertrophy, Angiotensin II, METTL3, miR-221/222, Wnt/β-catenin signaling
Address and Contact Information Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, 87 Hunan road, Nanjing 210000, Jiangsu, People’s Republic of China
*Corresponding authore: magenshan@hotmail.com
Read full article at BMC

No.  56DOI: 10.1186/s11658-022-00359-z Volume 27 (2022) - 27:56
Title THERAPEUTIC UTILITY OF MESENCHYMAL STROMAL CELL (MSC)‐BASED APPROACHES IN CHRONIC NEURODEGENERATION: A GLIMPSE INTO UNDERLYING MECHANISMS, CURRENT STATUS, AND PROSPECTS
Authors Mohaddeseh Rahbaran1, Angelina Olegovna Zekiy2, Mahta Bahramali3, Mohammadsaleh Jahangir4, Mahsa Mardasi5, Delaram Sakhaei6, Lakshmi Thangavelu7, Navid Shomali8, Majid Zamani9, Ali Mohammadi10* and Negin Rahnama11*
Abstract Recently, mesenchymal stromal cell (MSC)-based therapy has become an appreciated therapeutic approach in the context of neurodegenerative disease therapy. Accordingly, a myriad of studies in animal models and also some clinical trials have evinced the safety, feasibility, and efcacy of MSC transplantation in neurodegenerative conditions, most importantly in Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington’s disease (HD). The MSC-mediated desired efect is mainly a result of secretion of immunomodulatory factors in association with release of various neurotrophic factors (NTFs), such as glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Thanks to the secretion of protein-degrading molecules, MSC therapy mainly brings about the degradation of pathogenic protein aggregates, which is a typical appearance of chronic neurodegenerative disease. Such molecules, in turn, diminish neuroinfammation and simultaneously enable neuroprotection, thereby alleviating disease pathological symptoms and leading to cognitive and functional recovery. Also, MSC diferentiation into neural-like cells in vivo has partially been evidenced. Herein, we focus on the therapeutic merits of MSCs and also their derivative exosome as an innovative cell-free approach in AD, HD, PD, and ALS conditions. Also, we give a brief glimpse into novel approaches to potentiate MSC-induced therapeutic merits in such disorders, most importantly, administration of preconditioned MSCs.
Keywords Mesenchymal stromal cells (MSCs), Neurotrophic factors (NTFs), Neuroprotection, Diferentiation, Neuroinfammation
Address and Contact Information 1 Biotechnology Department, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
2 Department of Prosthetic Dentistry, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
3 Biotechnology Department, University of Tehran, Tehran, Iran.
4 Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
5 Biotechnology Department, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
6 School of Medicine, Sari Branch, Islamic Azad University, Sari, Iran.
7 Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India.
8 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
9 Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran.
10 Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran.
11 Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran.
*Corresponding author: Alimohammadi.neuro@gmail.com; rahnamanegin049@gmail.com
Read full article at BMC

No.  57DOI: 10.1186/s11658-022-00361-5 Volume 27 (2022) - 27:57
Title MOLECULAR MECHANISMS UNDERLYING THE RENAL PROTECTIVE EFECTS OF COENZYME Q10 IN ACUTE KIDNEY INJURY
Authors Shankun Zhao1†, Weizhou Wu2†, Jian Liao3†, Xinsheng Zhang1, Maolei Shen1, Xin Li1, Qi Lin1 and Chaoliang Cao4*
Abstract Coenzyme Q10 (CoQ10), an endogenous antioxidant, has been reported frequently to exert an outstanding protective efect on multiple organ injury, including acute kidney injury (AKI). In this study, we aim to summarize all the current evidence of the protective action of CoQ10 against AKI as there are presently no relevant reviews in the literature. After a systematic search, 20 eligible studies, either clinical trials or experimental studies, were included and further reviewed. CoQ10 treatment exhibited a potent renal protective efect on various types of AKI, such as AKI induced by drugs (e.g., ochratoxin A, cisplatin, gentamicin, L-NAME, and nonsteroidal anti-infammatory drug), extracorporeal shock wave lithotripsy (ESWL), sepsis, contrast media, and ischemia–reperfusion injury. The renal protective role of CoQ10 against AKI might be mediated by the antiperoxidative, anti-apoptotic, and anti-infammatory potential of CoQ10. The molecular mechanisms for the protective efects of CoQ10 might be attributed to the regulation of multiple essential genes (e.g., caspase-3, p53, and PON1) and signaling cascades (e.g., Nrf2/HO-1 pathway). This review highlights that CoQ10 may be a potential strategy in the treatment of AKI.
Keywords Coenzyme Q10, Acute kidney injury, Protection, Mechanism, Antioxidant
Address and Contact Information 1 Department of Urology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang, China
2 Department of Urology, Maoming People’s Hospital, Maoming 525000, Guangdong, China
3 Department of Nephrology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing 314001, Zhejiang, China
4 Department of Emergency Medicine, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang Province, China
Shankun Zhao, Weizhou Wu, and Jian Liao have contributed equally to this work
*Corresponding author: chaoliangcao2019@126.com
Read full article at BMC

No.  58DOI: 10.1186/s11658-022-00356-2 Volume 27 (2022) - 27:58
Title THE PROMISING THERAPEUTIC EFECTS OF METFORMIN ON METABOLIC REPROGRAMMING OF CANCER‐ASSOCIATED FBROBLASTS IN SOLID TUMORS
Authors Samaneh Mostafavi1*, Hamidreza Zalpoor2,3 and Zuhair Mohammad Hassan1*
Abstract Tumor-infltrated lymphocytes are exposed to many toxic metabolites and molecules in the tumor microenvironment (TME) that suppress their anti-tumor activity. Toxic metabolites, such as lactate and ketone bodies, are produced mainly by catabolic cancer-associated fbroblasts (CAFs) to feed anabolic cancer cells. These catabolic and anabolic cells make a metabolic compartment through which high-energy metabolites like lactate can be transferred via the monocarboxylate transporter channel 4. Moreover, a decrease in molecules, including caveolin-1, has been reported to cause deep metabolic changes in normal fbroblasts toward myofbroblast diferentiation. In this context, metformin is a promising drug in cancer therapy due to its efect on oncogenic signal transduction pathways, leading to the inhibition of tumor proliferation and downregulation of key oncometabolites like lactate and succinate. The cross-feeding and metabolic coupling of CAFs and tumor cells are also afected by metformin. Therefore, the importance of metabolic reprogramming of stromal cells and also the pivotal efects of metformin on TME and oncometabolites signaling pathways have been reviewed in this study.
Keywords Tumor microenvironment, Myofbroblasts, Cancer-associated fbroblasts, Metformin, Lactic acid, Stromal cells
Address and Contact Information 1 Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, 14115-154, Tehran, Iran
2 Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
3 Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientifc Education and Research Network (USERN), Tehran, Iran
*Corresponding author: samaneh.s.mostafavi@gmail.com; hasan_zm@modares.ac.ir
Read full article at BMC

No.  59DOI: 10.1186/s11658-022-00358-0 Volume 27 (2022) - 27:59
Title DNA METHYLATION‐MEDIATED DIFERENTIAL EXPRESSION OF DLX4 ISOFORMS HAS OPPOSING ROLES IN LEUKEMOGENESIS
Authors Jing‐dong Zhou1,2,3†, Yang‐jing Zhao4†, Jia‐yan Leng1,2,3†, Yu Gu1,2,3, Zi‐jun Xu2,3,5, Ji‐chun Ma2,3,5, Xiang‐mei Wen2,3,5, Jiang Lin2,3,5*, Ting‐juan Zhang2,3,6* and Jun Qian1,2,3*
Abstract Background: Previously, we reported the expression of DLX4 isoforms (BP1 and DLX7) in myeloid leukemia, but the functional role of DLX4 isoforms remains poorly understood. In the work described herein, we further determined the underlying role of DLX4 isoforms in chronic myeloid leukemia (CML) leukemogenesis.
Methods: The expression and methylation of DLX4 isoforms were detected by realtime quantitative PCR (RT-qPCR) and real-time quantitative methylation-specifc PCR (RT-qMSP) in patients with CML. The functional role of DLX4 isoforms was determined in vitro and in vivo. The molecular mechanism of DLX4 isoforms in leukemogenesis was identifed based on chromatin immunoprecipitation with high-throughput sequencing (ChIP-Seq)/assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-Seq) and RNA sequencing (RNA-Seq).
Results: BP1 expression was increased in patients with CML with unmethylated promoter, but DLX7 expression was decreased with hypermethylated promoter. Functionally, overexpression of BP1 increased the proliferation rate of K562 cells with S/G2 promotion, whereas DLX7 overexpression reduced the proliferation rate of K562 cells with G1 arrest. Moreover, K562 cells with BP1 overexpression increased the tumorigenicity in NCG mice, whereas K562 cells with DLX7 overexpression decreased the tumorigenicity. Mechanistically, a total of 91 genes including 79 messenger RNAs (mRNAs) and 12 long noncoding RNAs (lncRNAs) were discovered by ChIP-Seq and RNA-Seq as direct downstream targets of BP1. Among the downstream genes, knockdown of RREB1 and SGMS1-AS1 partially revived the proliferation caused by BP1 overexpression in K562 cells. Similarly, using ATAC-Seq and RNA-Seq, a total of 282 genes including 151 mRNA and 131 lncRNAs were identifed as direct downstream targets of DLX7. Knockdown of downstream genes PTPRB and NEAT1 partially revived the proliferation caused by DLX7 overexpression in K562 cells. Finally, we also identifed and validated a SGMS1-AS1/miR-181d-5p/SRPK2 competing endogenous RNA (ceRNA) network caused by BP1 overexpression in K562 cells.
Conclusions: The current fndings reveal that DNA methylation-mediated diferential expression of DLX4 isoforms BP1 and DLX7 plays opposite functions in leukemogenesis. BP1 plays an oncogenic role in leukemia development, whereas DLX7 acts as a tumor suppressor gene. These results suggest DLX4 as a therapeutic target for antileukemia therapy.
Keywords DLX4, Expression, Methylation, Function, Leukemogenesis
Address and Contact Information 1 Department of Hematology, Afliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang 212002, Jiangsu, People’s Republic of China
2 Zhenjiang Clinical Research Center of Hematology, Zhenjiang 212002, Jiangsu, People’s Republic of China
3 The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Zhenjiang 212002, Jiangsu, People’s Republic of China
4 Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, People’s Republic of China
5 Laboratory Center, Afliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang 212002, Jiangsu, People’s Republic of China
6 Department of Oncology, Afliated People’s Hospital of Jiangsu University, 8 Dianli Rd., Zhenjiang 212002, Jiangsu, People’s Republic of China
*Corresponding author: linjiangmail@qq.com; zhangtingjuan1990@qq.com; qianjun@ujs.edu.cn
† Jing-dong Zhou, Yang-jing Zhao, and Jia-yan Leng contributed equally to this work
Read full article at BMC

No.  60DOI: 10.1186/s11658-022-00355-3 Volume 27 (2022) - 27:60
Title QUERCETIN AS A JAK–STAT INHIBITOR: A POTENTIAL ROLE IN SOLID TUMORS AND NEURODEGENERATIVE DISEASES
Authors Hamidreza Zalpoor1,2, Mohsen Nabi‐Aadi3, Razieh Forghaniesfidvajani2, Chanour Tavakol4, Faranak Farahighasreaboonasr5, Farid Pakizeh6, Vahid Ghobadi Dana7 and Farhad Seif7,8*
Abstract The Janus kinase–signal transducer and activator of transcription (JAK–STAT) pathway is involved in many immunological processes, including cell growth, proliferation, diferentiation, apoptosis, and infammatory responses. Some of these processes can contribute to cancer progression and neurodegeneration. Owing to the complexity of this pathway and its potential crosstalk with alternative pathways, monotherapy as targeted therapy has usually limited long-term efcacy. Currently, the majority of JAK–STAT-targeting drugs are still at preclinical stages. Meanwhile, a variety of plant polyphenols, especially quercetin, exert their inhibitory efects on the JAK–STAT pathway through known and unknown mechanisms. Quercetin has shown prominent inhibitory efects on the JAK–STAT pathway in terms of anti-infammatory and antitumor activity, as well as control of neurodegenerative diseases. This review discusses the pharmacological efects of quercetin on the JAK–STAT signaling pathway in solid tumors and neurodegenerative diseases.
Keywords Quercetin, JAK–STAT inhibitor, Solid tumors, Neurodegenerative diseases, Cancers
Address and Contact Information 1 Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
2 Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
3 Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
4 Tehran University of Medical Sciences, Tehran, Iran
5 Department of Biology, Zand Institute of Higher Education, Shiraz, Iran
6 Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
7 Department of Immunology and Allergy, Academic Center for Education, Culture, and Research (ACECR), Tehran, Iran
8 Neuroscience Research Center, Iran University of Medical Sciences, Enghelab St., Aboureyhan St., Vahid Nazari Crossroad, P17, Tehran Postal code: 1315795613, Iran
*Corresponding author: farhad.seif@outlook.com
Read full article at BMC

No.  61DOI: 10.1186/s11658-022-00362-4 Volume 27 (2022) - 27:61
Title d‐BORNEOL ENHANCES CISPLATIN SENSITIVITY VIA p21/p27‐MEDIATED S‐PHASE ARREST AND CELL APOPTOSIS IN NON‐SMALL CELL LUNG CANCER CELLS AND A MURINE XENOGRAFT MODEL
Authors Jinxiu Li1, Jianmei Yuan1, Yong Li1, Jian Wang1*, Daoyin Gong2*, Qian Xie1, Rong Ma1, Jiajun Wang1, Mihong Ren1, Danni Lu1 and Zhuo Xu1
Abstract Background: Cisplatin (CDDP) is commonly used to treat non-small cell lung cancer (NSCLC), but the appearance of drug resistance greatly hinders its efcacy. Borneol may promote drug absorption; however, synergism between borneol and CDDP in suppressing NSCLC is not clearly understood. Hence, we investigated borneol as a novel chemosensitizer to support chemotherapeutic efcacy and reduce side efects.
Methods: We compared viability after exposure to d-borneol, l-borneol, and synthetic borneol in two NSCLC cell lines, A549 and H460, and selected the most sensitive cells. We then assessed synergy between borneol forms and CDDP in cisplatin-resistant NSCLC cells, H460/CDDP. Next, we identifed efective concentrations and exposure times. Subsequently, we evaluated cell migration via wound healing and cell proliferation via clone formation assay. Then, we focused on P-glycoprotein (P-gp) function, cell cycle, apoptosis, and RNA sequencing to elucidate underlying molecular mechanisms for synergy. Finally, we used an H460/CDDP xenograft tumor model to verify antitumor activity and safety in vivo. Data were examined using one-way analysis of variance (ANOVA) for multiple datasets or t-test for comparisons between two variables.
Results: d-Borneol was more efective in H460 than A549 cells. d-Borneol combined with CDDP showed greater inhibition of cell proliferation, migration, and clone formation in H460/CDDP cells than CDDP alone. RNA sequencing (RNA-seq) analysis identifed diferentially expressed genes enriched in cell cycle pathways. The impact of d-borneol on CDDP chemosensitivity involved arrest of the cell cycle at S phase via p27/p21-mediated cyclinA2/D3-CDK2/6 signaling and activation of intrinsic apoptosis via p21-mediated Bax/Bcl-2/caspase3 signaling. Further, d-borneol ameliorated drug resistance by suppressing levels and activity of P-gp. Cotreatment with d-borneol and CDDP inhibited tumor growth in vivo and reduced CDDP-caused liver and kidney toxicity.
Conclusions: d-Borneol increased the efcacy of cisplatin and reduced its toxicity. This compound has the potential to become a useful chemosensitizer for drug-resistance NSCLC.
Keywords d-Borneol, Non-small cell lung cancer, Drug resistance, Cisplatin, Chemosensitizer, Cell cycle, Apoptosis
Address and Contact Information 1 State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
2 Department of Pathology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
*Corresponding author: lczyx712@163.com; daoyinggong@163.com
Read full article at BMC

No.  62DOI: 10.1186/s11658-022-00365-1 Volume 27 (2022) - 27:62
Title ARTEMISININ RELIEVES OSTEOARTHRITIS BY ACTIVATING MITOCHONDRIAL AUTOPHAGY THROUGH REDUCING TNFSF11 EXPRESSION AND INHIBITING PI3K/AKT/mTOR SIGNALING IN CARTILAGE
Authors Jin Li1†, Mengqing Jiang2†, Zhentang Yu3, Chenwei Xiong3, Jieen Pan1, Zhenhai Cai1, Nanwei Xu3, Xindie Zhou3*, Yong Huang3 and Zhicheng Yang3*
Abstract Osteoarthritis (OA) is a widespread chronic degenerative joint disease characterized by the degeneration of articular cartilage or infamed joints. Our fndings indicated that treatment with artemisinin (AT) downregulates the protein levels of MMP3, MMP13, and ADAMTS5, which are cartilage degradation-related proteins in OA, and inhibits the expression of infammatory factors in interleukin-1β (IL-1β)-stimulated chondrocytes. However, the mechanism of the role of AT in OA remains unclear. Here, we performed gene sequencing and bioinformatics analysis in control, OA, and OA+AT groups to demonstrate that several mRNA candidates were enriched in the PI3K/AKT/mTOR signaling pathway, and TNFSF11 was signifcantly downregulated after AT treatment. TNFSF11 was downregulated in the OA+AT group, whereas it was upregulated in rat OA tissues and OA chondrocytes. Therefore, we confrmed that TNFSF11 was the target gene of AT. In addition, our study revealed that AT relieved cartilage degradation and defection by activating mitochondrial autophagy via inhibiting the PI3K/AKT/mTOR signaling pathway in IL-1β-induced chondrocytes. Furthermore, an OA model was established in rats with medial meniscus destabilization. Injecting AT into the knee joints of OA rat alleviated surgical resection-induced cartilage destruction. Thus, these fndings revealed that AT relieves OA by activating mitochondrial autophagy by reducing TNFSF11 expression and inhibiting PI3K/AKT/mTOR signaling.
Keywords Artemisinin, PI3K/AKT signaling pathway, Autophagy, TNFSF11, Mitochondria, Osteoarthritis
Address and Contact Information 1 Department of Orthopedic Surgery, The Second Afliated Hospital of Jiaxing University, Jiaxing 314000, China
2 Department of Pharmacy, The Second Afliated Hospital of Jiaxing University, Jiaxing 314000, China
3 Department of Orthopedics, The Afliated Changzhou No.2 People’s Hospital of Nanjing Medical University, Changzhou 213000, China
*Corresponding author: zhouxindie@njmu.edu.cn; yangzhicheng@njmu.edu.cn
Jin Li and Mengqing Jiang contributed equally to this study
Read full article at BMC

No.  63DOI: 10.1186/s11658-022-00363-3 Volume 27 (2022) - 27:63
Title COMPREHENSIVE OVERVIEW OF COVID‐19‐RELATED RESPIRATORY FAILURE: FOCUS ON CELLULAR INTERACTIONS
Authors Fahimeh Zamani Rarani1, Mohammad Zamani Rarani1, Michael R. Hamblin2, Bahman Rashidi1*, Seyed Mohammad Reza Hashemian3* and Hamed Mirzaei4,5*
Abstract The pandemic outbreak of coronavirus disease 2019 (COVID-19) has created health challenges in all parts of the world. Understanding the entry mechanism of this virus into host cells is essential for efective treatment of COVID-19 disease. This virus can bind to various cell surface molecules or receptors, such as angiotensin-converting enzyme 2 (ACE2), to gain cell entry. Respiratory failure and pulmonary edema are the most important causes of mortality from COVID-19 infections. Cytokines, especially proinfammatory cytokines, are the main mediators of these complications. For normal respiratory function, a healthy air–blood barrier and sufcient blood fow to the lungs are required. In this review, we frst discuss airway epithelial cells, airway stem cells, and the expression of COVID-19 receptors in the airway epithelium. Then, we discuss the suggested molecular mechanisms of endothelial dysfunction and blood vessel damage in COVID-19. Coagulopathy can be caused by platelet activation leading to clots, which restrict blood fow to the lungs and lead to respiratory failure. Finally, we present an overview of the efects of immune and non-immune cells and cytokines in COVID-19-related respiratory failure.
Keywords SARS-CoV-2, COVID-19, Airway epithelial cells, Endothelial cells, Platelets, Cytokines, Pulmonary edema
Address and Contact Information 1 Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
2 Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
3 Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
4 Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
5 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, IR, Iran
*Corresponding author: b_rashidi@med.mui.ac.ir; smrhashemian@sbmu.ac.ir; hashemian.smr4r4@gmil.com; mirzaei-h@kaums.ac.ir; h.mirzaei2002@gmail.com
Read full article at BMC

No.  64DOI: 10.1186/s11658-022-00360-6 Volume 27 (2022) - 27:64
Title GRANULIN AS AN IMPORTANT IMMUNE MOLECULE INVOLVED IN LAMPREY TISSUE REPAIR AND REGENERATION BY PROMOTING CELL PROLIFERATION AND MIGRATION
Authors Ruixiang Sun1,2,3†, Dong Wang1,2,3†, Yuxuan Song1,2,3†, Qingwei Li1,2,3, Peng Su1,2,3* and Yue Pang1,2,3*
Abstract Progranulin (PGRN) is an autocrine growth factor that regulates cell proliferation, migration, wound healing, and tissue repair in mammals. Lamprey is the most primitive of the extant vertebrates and is regarded as the survivor of a once fourishing group of paleozoic vertebrates, with a history of more than 500 million years. To date, the evolutionary dynamics and the underlying function of the PGRNs remain largely unclear in lamprey. Here, we screened four genes encoding PGRNs from the genomes of Lethenteron reissneri and Petromyzon marinus, including one long form (named Lr-PGRN-L) and three short forms (named Lr-PGRN-S1, Lr-PGRN-S2, and Lr-PGRN-S3), and performed phylogenetic tree, functional domain, and synteny analyses to identify the evolutionary history of the four Lr-PGRNs. In addition, the expressions of the four Lr-pgrn family genes and the immune response against various pathogenic challenges were also investigated. We found that these genes were widely distributed in various tissues of lamprey and performed a variety of functions. Moreover, our results suggest that Lr-PGRN-S1 induces cell migration and proliferation, and is involved in repair after skin and spinal cord injury under appropriate conditions. Our fndings are valuable because they improve the understanding of the evolutionary relationship of vertebrate pgrn genes, as well as providing new insights into the diverse and important roles of Lr-PGRNs.
Keywords PGRN, Lamprey, Immune response, Tissue repair
Address and Contact Information 1 College of Life Sciences, Liaoning Normal University, Dalian 116081, China
2 Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
3 Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
*Corresponding author: sp4046@163.com; pangyue01@163.com
Ruixiang Sun, Dong Wang, and Yuxuan Song contributed equally to this work
Read full article at BMC

No.  65DOI: 10.1186/s11658-022-00354-4 Volume 27 (2022) - 27:65
Title RECENT INSIGHTS INTO THE microRNA‐DEPENDENT MODULATION OF GLIOMAS FROM PATHOGENESIS TO DIAGNOSIS AND TREATMENT
Authors Alireza Maf1†, Atefe Rahmati2,3†, Zahra Babaei Aghdam4, Raziyeh Salami5, Marziyeh Salami6, Omid Vakili1* and Esmat Aghadavod7,8*
Abstract Gliomas are the most lethal primary brain tumors in adults. These highly invasive tumors have poor 5-year survival for patients. Gliomas are principally characterized by rapid difusion as well as high levels of cellular heterogeneity. However, to date, the exact pathogenic mechanisms, contributing to gliomas remain ambiguous. MicroRNAs (miRNAs), as small noncoding RNAs of about 20 nucleotides in length, are known as chief modulators of diferent biological processes at both transcriptional and posttranscriptional levels. More recently, it has been revealed that these noncoding RNA molecules have essential roles in tumorigenesis and progression of multiple cancers, including gliomas. Interestingly, miRNAs are able to modulate diverse cancer-related processes such as cell proliferation and apoptosis, invasion and migration, diferentiation and stemness, angiogenesis, and drug resistance; thus, impaired miRNAs may result in deterioration of gliomas. Additionally, miRNAs can be secreted into cerebrospinal fuid (CSF), as well as the bloodstream, and transported between normal and tumor cells freely or by exosomes, converting them into potential diagnostic and/or prognostic biomarkers for gliomas. They would also be great therapeutic agents, especially if they could cross the blood–brain barrier (BBB). Accordingly, in the current review, the contribution of miRNAs to glioma pathogenesis is frst discussed, then their glioma-related diagnostic/prognostic and therapeutic potential is highlighted briefy.
Keywords Glioma, Brain neoplasms, MicroRNAs, Carcinogenesis, Biomarkers, Therapeutics
Address and Contact Information 1 Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
2 Department of Hematology and Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
3 Department of Basic Science, Neyshabur University of Medical Science, Neyshabur, Iran.
4 Imaging Sciences Research Group, Tabriz University of Medical Sciences, Tabriz, Iran.
5 Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
6 Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
7 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
8 Department of Clinical Biochemistry, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
*Corresponding: o.vakili.isf@gmail.com; aghadavodm@gmail.com
Alireza Maf and Atefe Rahmati contributed equally to this study, and both are frst authors.
Read full article at BMC

No.  66DOI: 10.1186/s11658-022-00367-z Volume 27 (2022) - 27:66
Title SESN2 PREVENTS THE SLOW‐TO‐FAST MYOFBER SHIFT IN DENERVATED ATROPHY VIA AMPK/PGC‐1α PATHWAY
Authors Xiaofan Yang1†, Pingping Xue2, Zhenyu Liu1, Wenqing Li3, Chuyan Li3 and Zhenbing Chen1*
Abstract Background: Sestrin2 (SESN2), a stress-inducible protein, has been reported to protect against denervated muscle atrophy through unfolded protein response and mitophagy, while its role in myofber type transition remains unknown.
Methods: A mouse sciatic nerve transection model was created to evaluate denervated muscle atrophy. Myofber type transition was confrmed by western blot, fuorescence staining, ATP quantifcation, and metabolic enzyme activity analysis. Adeno-associated virus (AAV) was adopted to achieve SESN2 knockdown and overexpression in gastrocnemius. AMPK/PGC-1α signal was detected by western blot and activated with 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR). C2C12 myotubes with rotenone treatment were adopted for in vitro experiments.
Results: SESN2 was found to be upregulated in denervated skeletal muscles and rotenone-treated C2C12 cells. Knockdown of SESN2 aggravated muscle atrophy and accelerated myofber type transition from slow-twitch to fast-twitch. Moreover, AMPK/PGC-1α signaling was proven to be activated by SESN2 after denervation, which further induced the expression of hypoxia-inducible factor HIF2α. Exogenous activation of AMPK/PGC-1α signaling could counteract the addition of slow-to-fast myofber shift caused by SESN2 knockdown and lead to the retainment of muscle mass after denervation.
Conclusion: Collectively, the present study indicates that SESN2 prevents myofber type transition from slow-twitch to fast-twitch and preserves muscle mass in denervated atrophy via AMPK/PGC-1α signaling. These fndings contribute to a better understanding of the pathogenesis of muscle atrophy and provide novel insights into the role of SESN2 in myofber type transition.
Keywords Denervation, Skeletal muscle atrophy, Myofber type transition, SESN2, AMPK/PGC-1α
Address and Contact Information 1 Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
2 Department of Pharmacy, Tongji Hospital, Tongji Medica College, Huazhong University of Science and Technology, Wuhan 430030, China
3 Department of Hand and Foot Surgery, Union Shenzhen Hospital, Huazhong University of Science and Technology, Shenzhen 518052, China
*Corresponding author: zbchen@hust.edu.cn
Xiaofan Yang and Pingping Xue contributed equally to this work.
Read full article at BMC

No.  67DOI: 10.1186/s11658-022-00369-x Volume 27 (2022) - 27:67
Title RETRACTION NOTE TO: PARECOXIB INHIBITS ESOPHAGEAL SQUAMOUS CELL CARCINOMA PROGRESSION VIA THE PDK1–AKT PATHWAY
Authors Han‐Ming Huang1, Xiao‐Yu Huang1, Shao‐Ping Wu1, Can‐Keng Chen1, Xin‐Hua He2* and Yong‐Fa Zhang1*
Abstract Retraction to: Cellular & Molecular Biology Letters (2022) 27:28
https://doi.org/10.1186/s11658-022-00324-w


The Editor-in-Chief has retracted this article at the authors’ request. After publication, the authors became aware that the KYSE180 cells used in this study were cross-contaminated. Further checks by the publisher identifed high similarity between the KYSE180 200 and 300 μM middle images. Additionally, the authors have stated that the number of injected cells for the in vivo tumor growth assay provided in the Materials and methods is incorrect.

All authors agree to this retraction.


Published online: 15 August 2022


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 Anesthesiology, Second Afliated Hospital of Shantou University Medical College, Shantou 515041, People’s Republic of China
2 Department of Physiology, Shantou University Medical College, Shantou 515041, People’s Republic of China
*Corresponding author: Hexh@stu.edu.cn; 10yfzhang1@stu.edu.cn
The original article can be found online at https://doi.org/10.1186/s11658-​022-​00324-w.
Read full article at BMC

No.  68DOI: 10.1186/s11658-022-00373-1 Volume 27 (2022) - 27:68
Title CORRECTION: d‐BORNEOL ENHANCES CISPLATIN SENSITIVITY VIA p21/p27‐MEDIATED S‐PHASE ARREST AND CELL APOPTOSIS IN NON‐SMALL CELL LUNG CANCER CELLS AND A MURINE XENOGRAFT MODEL
Authors Jinxiu Li1, Jianmei Yuan1, Yong Li1, Jian Wang1*, Daoyin Gong2*, Qian Xie1, Rong Ma1, Jiajun Wang1, Mihong Ren1, Danni Lu1 and Zhuo Xu1
Abstract Correction: Cellular & Molecular Biology Letters (2022) 27:61
https://doi.org/10.1186/s11658-022-00362-4


Following publication of the original article [1], the authors informed us that would like to replace the Figure 1L. Te correct Fig. 1 is given in link below:

Also, an error was identifed in the Results section.
Te updated phrase is given below and the changes have been highlighted in bold typeface.
Results
d‐Borneol combined with CDDP suppresses NSCLC tumor growth in vivo

We noted that treatment with d-borneol alone caused no notable body weight change, but the body weight of animals treated with CDDP was signifcantly lower than that of those in the Con group. After 14 days of treatment with the combination of d-borneol and CDDP, the CDDP+Bor group increased body weight signifcantly compared with the CDDP group, indicating that d-borneol weaken the toxicity of cisplatin in vivo (Fig. 6E) 0.2111.
Te original article has been corrected.

Published online: 18 August 2022


Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional afliations.
Keywords
Address and Contact Information 1 State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
2 Department of Pathology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
*Corresponding author: lczyx712@163.com; daoyinggong@163.com
The original article can be found online at https://doi.org/10.1186/s11658-022-00362-4.
Read full article at BMC

No.  69DOI: 10.1186/s11658-022-00366-0 Volume 27 (2022) - 27:69
Title AGING OF MESENCHYMAL STEM CELL: MACHINERY, MARKERS, AND STRATEGIES OF FiGHTING
Authors Mahmoud Al‐Azab1*†, Mohammed Saf2†, Elina Idiiatullina1,3†, Fadhl Al‐Shaebi2 and Mohamed Y. Zaky4
Abstract Human mesenchymal stem cells (MSCs) are primary multipotent cells capable of diferentiating into osteocytes, chondrocytes, and adipocytes when stimulated under appropriate conditions. The role of MSCs in tissue homeostasis, aging-related diseases, and cellular therapy is clinically suggested. As aging is a universal problem that has large socioeconomic efects, an improved understanding of the concepts of aging can direct public policies that reduce its adverse impacts on the healthcare system and humanity. Several studies of aging have been carried out over several years to understand the phenomenon and diferent factors afecting human aging. A reduced ability of adult stem cell populations to reproduce and regenerate is one of the main contributors to the human aging process. In this context, MSCs senescence is a major challenge in front of cellular therapy advancement. Many factors, ranging from genetic and metabolic pathways to extrinsic factors through various cellular signaling pathways, are involved in regulating the mechanism of MSC senescence. To better understand and reverse cellular senescence, this review highlights the underlying mechanisms and signs of MSC cellular senescence, and discusses the strategies to combat aging and cellular senescence.
Keywords Aging, Diferentiation, Mesenchymal stem cell, Cellular senescence, Senescence markers; Anti-cellular senescence
Address and Contact Information 1 Department of Immunology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
2 Respiratory Diseases, Shandong Second Provincial General Hospital, Shandong University, Jinan, China 3 Department of Therapy and Nursing, Bashkir State Medical University, 450008 Ufa, Russia
4 Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
*Corresponding author: azab.m12@gmail.com
Mahmoud Al-Azab, Mohammed Saf, and Elina Idiiatullina contributed equally to this work.
Read full article at BMC

No.  70DOI: 10.1186/s11658-022-00368-y Volume 27 (2022) - 27:70
Title THE CURRENT LANDSCAPE OF microRNAs (miRNAs) IN BACTERIAL PNEUMONIA: OPPORTUNITIES AND CHALLENGES
Authors Fan Zhang, Yunxin Zhou and Junying Ding*
Abstract MicroRNAs (miRNAs), which were initially discovered in Caenorhabditis elegans, can regulate gene expression by recognizing cognate sequences and interfering with the transcriptional or translational machinery. The application of bioinformatics tools for structural analysis and target prediction has largely driven the investigation of certain miRNAs. Notably, it has been found that certain miRNAs which are widely involved in the infammatory response and immune regulation are closely associated with the occurrence, development, and outcome of bacterial pneumonia. It has been shown that certain miRNA techniques can be used to identify related targets and explore associated signal transduction pathways. This enhances the understanding of bacterial pneumonia, notably for “refractory” or drug-resistant bacterial pneumonia. Although these miRNA-based methods may provide a basis for the clinical diagnosis and treatment of this disease, they still face various challenges, such as low sensitivity, poor specifcity, low silencing efciency, of-target efects, and toxic reactions. The opportunities and challenges of these methods have been completely reviewed, notably in bacterial pneumonia. With the continuous improvement of the current technology, the miRNA-based methods may surmount the aforementioned limitations, providing promising support for the clinical diagnosis and treatment of “refractory” or drug-resistant bacterial pneumonia.
Keywords miRNA, Bacterial pneumonia, Host–pathogen, Sensitivity, Specifcity, Offtarget efect
Address and Contact Information Beijing Key Laboratory of Basic Research With Traditional Chinese Medicine On Infectious Diseases, Beijing Institute of Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
*Corresponding author: 18211197728@163.com
Read full article at BMC

No.  71DOI: 10.1186/s11658-022-00378-w Volume 27 (2022) - 27:71
Title THE O‐GLYCOSYLATING ENZYME GALNT2 ACTS AS AN ONCOGENIC DRIVER IN NON‐SMALL CELL LUNG CANCER
Authors Qing Hu1,2†, Tian Tian2†, Yahui Leng2†, Yuanhui Tang1, Shuang Chen2, Yueyao Lv2, Jingyin Liang2, Yanni Liu2, Tianhui Liu2, Li Shen1,2* and Xiaoxia Dong1,2*
Abstract Background: N-Acetylgalactosaminyltransferases (GALNTs), the enzymes that initiate mucin-type O-glycosylation, are closely associated with tumor occurrence and progression. However, a comprehensive analysis of GALNTs in non-small cell lung cancer (NSCLC) is lacking.
Methods: The expression profles and prognostic values of the GALNT family members in NSCLC were analyzed using publicly available databases. Gain- and loss-of-function experiments were applied to assess the biological function of GALNT2 in NSCLC. High-throughput sequencing and bioinformatics approaches were employed to uncover the regulatory mechanism of GALNT2.
Results: Among the family members of GALNTs, only GALNT2 was frequently overexpressed in NSCLC tissues and was positively correlated with poor prognosis. In vitro assays showed that GALNT2 knockdown repressed NSCLC cell proliferation, migration, and invasion, but induced apoptosis and cell cycle arrest. Correspondently, GALNT2 overexpression exerted the opposite efects. In vivo experiments demonstrated that knockdown of GALNT2 restrained tumor formation in nude mice. Mechanistic investigations revealed that GALNT2 modifed the O-glycosylation of ITGA5 and afected the activation of the PI3K/Akt and MAPK/ERK pathways. Further studies showed that miR-30d was a negative regulator of GALNT2.
Conclusions: These fndings suggest that GALNT2 is an oncogene in NSCLC and has the potential as a target for NSCLC therapy.
Keywords Non-small cell lung cancer, Oncogene, Glycosyltransferase, GALNT2
Address and Contact Information 1 Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China
2 Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
*Corresponding author: shenlihb@163.com; xiaoxia.28@163.com
Qing Hu, Tian Tian and Yahui Leng contributed equally to this article
Read full article at BMC

No.  72DOI: 10.1186/s11658-022-00371-3 Volume 27 (2022) - 27:72
Title OSTEOPOROSIS PATHOGENESIS AND TREATMENT: EXISTING AND EMERGING AVENUES
Authors Bo Liang1, George Burley2, Shu Lin2,3*† and Yan‐Chuan Shi2,3,4*†
Abstract Osteoporotic fractures lead to increased disability and mortality in the elderly population. With the rapid increase in the aging population around the globe, more efective treatments for osteoporosis and osteoporotic fractures are urgently required. The underlying molecular mechanisms of osteoporosis are believed to be due to the increased activity of osteoclasts, decreased activity of osteoblasts, or both, which leads to an imbalance in the bone remodeling process with accelerated bone resorption and attenuated bone formation. Currently, the available clinical treatments for osteoporosis have mostly focused on factors infuencing bone remodeling; however, they have their own limitations and side efects. Recently, cytokine immunotherapy, gene therapy, and stem cell therapy have become new approaches for the treatment of various diseases. This article reviews the latest research on bone remodeling mechanisms, as well as how this underpins current and potential novel treatments for osteoporosis.
Keywords Osteoporosis, Pathogenesis, Bone remodeling, Bone formation, Bone resorption, MicroRNA-based therapy, Stem cell therapy
Address and Contact Information 1 Department of Endocrinology and Metabolism, The Second Afliated Hospital of Fujian Medical University, Quanzhou, China
2 Neuroendocrinology Group, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
3 Centre of Neurological and Metabolic Research, The Second Afliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou 362000, Fujian Province, China
4 St Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, Australia
*Corresponding author: shulin1956@126.com; y.shi@garvan.org.au
Shu Lin and Yan-Chuan Shi are joint senior authors.
Read full article at BMC

No.  73DOI: 10.1186/s11658-022-00375-z Volume 27 (2022) - 27:73
Title MELATONIN AND CANCER SUPPRESSION: INSIGHTS INTO ITS EFECTS ON DNA METHYLATION
Authors Amirhossein Davoodvandi1,2, Banafsheh Nikfar3, Russel J. Reiter4 and Zatollah Asemi5*
Abstract Melatonin is an important naturally occurring hormone in mammals. Melatonin-mediated biological efects include the regulation of circadian rhythms, which is important for optimal human health. Also, melatonin has a broad range of immunoenhancing actions. Moreover, its oncostatic properties, especially regarding breast cancer, involve a variety cancer-inhibitory processes and are well documented. Due to their promising efects on the prognosis of cancer patients, anti-cancer drugs with epigenetic actions have attracted a signifcant amount of attention in recent years. Epigenetic modifcations of cancers are categorized into three major processes including non-coding RNAs, histone modifcation, and DNA methylation. Hence, the modifcation of the latter epigenetic event is currently considered an efective strategy for treatment of cancer patients. Thereby, this report summarizes the available evidence that investigated melatonin-induced efects in altering the status of DNA methylation in diferent cancer cells and models, e.g., malignant glioma and breast carcinoma. Also, we discuss the role of artifcial light at night (ALAN)-mediated inhibitory efects on melatonin secretion and subsequent impact on global DNA methylation of cancer cells.
Keywords Melatonin, DNA methylation, DNMT, Epigenetics
Address and Contact Information 1 Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
2 Cancer Immunology Project (CIP), Universal Scientifc Education and Research Network (USERN), Tehran, Iran
3 Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
4 Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA
5 Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
*Corresponding author: Zatollah Asemi Asemi_r@yahoo.com
Read full article at BMC

No.  74DOI: 10.1186/s11658-022-00377-x Volume 27 (2022) - 27:74
Title EXOSOME APPLICATION IN TREATMENT AND DIAGNOSIS OF B-CELL DISORDERS: LEUKEMIAS, MULTIPLE SCLEROSIS, AND ARTHRITIS RHEUMATOID
Authors Mohsen Karami Fath1, Jalil Azami2, Niloofar Jaafari3, Mahsa Akbari Oryani4, Nafseh Jafari5, Alireza Karim poor6, Ali Azargoonjahromi7, Mohsen Nabi-Afadi8, Zahra Payandeh9,12*, Hamidreza Zalpoor10,11* and Dariush Shanehbandi9,12*
Abstract Exosomes, known as a type of extracellular vesicles (EVs), are lipid particles comprising heterogeneous contents such as nucleic acids, proteins, and DNA. These bi-layered particles are naturally released into the extracellular periphery by a variety of cells such as neoplastic cells. Given that exosomes have unique properties, they can be used as vectors and carriers of biological and medicinal particles like drugs for delivering to the desired areas. The proteins and RNAs being encompassed by the circulating exosomes in B-cell malignancies are deemed as the promising sources for diagnostic and prognostic biomarkers, as well as therapeutic agents. Exosomes can also provide a “snapshot” view of the tumor and metastatic landscape at any particular time. Further, clinical research has shown that exosomes are produced by immune cells such as dendritic cells can stimulate the immune system, so these exosomes can be used in antitumor vaccines. Despite the great potential of exosomes in the felds of diagnostic and treatment, further studies are in need for these purposes to reach a convergence notion. This review highlights the applications of exosomes in multiple immune-related diseases, including chronic lymphocytic leukemia, multiple sclerosis, and arthritis rheumatoid, as well as explaining sundry aspects of exosome therapy and the function of exosomes in diagnosing diseases.
Keywords Exosome, Cancer, Chronic lymphocytic leukemia, Acute myeloid leukemia, Rheumatic arthritis, Multiple sclerosis
Address and Contact Information 1 Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
2 Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
3 Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
4 Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
5 Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
6 School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
7 Shiraz University of Medical Sciences, Shiraz, Iran
8 Department of Biochemistry, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
9 Department Medical Biochemistry and Biophysics, Division Medical Infammation Research, Karolinska Institute, Stockholm, Sweden
10 Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
11 Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientifc Education & Research Network (USERN), Tehran, Iran
12 Immunology Research center, Tabriz University of Medical Science, Tabriz, Iran
*Corresponding author: Zahra Payandeh Zpayandeh58@yahoo.com
Read full article at BMC

No.  75DOI: 10.1186/s11658-022-00374-0 Volume 27 (2022) - 27:75
Title Pou3f1 MEDIATES THE EFECT OF Nfatc3 ON ULCERATIVE COLITIS-ASSOCIATED COLORECTAL CANCER BY REGULATING INFAMMATION
Authors Yan Lin1, Dongxu Wang1, Hong Zhao1,2, Dongyue Li1,3, Xinning Li1,4 and Lianjie Lin1*
Abstract Background: Ulcerative colitis-associated colorectal cancer (UC-CRC) is an important complication of ulcerative colitis. Pou3f1 (POU class 3 homeobox 1) is a critical regulator for developmental events and cellular biological processes. However, the role of Pou3f1 in the development of UC-CRC is unclear.
Methods: In vivo, a UC-CRC mouse model was induced by azoxymethane (AOM) and dextran sulfate sodium (DSS). Body weight, colon length, mucosal damage, tumor formation, and survival rate were assessed to determine the progression of UC-CRC. Western blot, quantitative real-time PCR, ELISA, immunohistochemistry, immunofuorescence and TUNEL were performed to examine the severity of infammation and tumorigenesis. In vitro, LPS-treated mouse bone marrow-derived macrophages (BMDMs) and RAW264.7 cells were used to study the role of Pou3f1 in infammation. ChIP and luciferase reporter assays were used to confrm the interaction between Nfatc3 and Pou3f1.
Results: Pou3f1 expression was increased in the colons of UC-CRC mice, and its inhibition attenuated mucosal injury, reduced colon tumorigenesis and increased survival ratio. Knockdown of Pou3f1 suppressed cell proliferation and increased cell death in colon tumors. Both the in vivo and in vitro results showed that Pou3f1 depletion reduced the production of proinfammation mediators. In addition, ChIP and luciferase reporter assays demonstrated that Nfatc3 directly bound with the Pou3f1 promoter to induce its expression. The efect of Nfatc3 on the infammatory response in macrophages was suppressed by Pou3f1 knockdown.
Conclusion: Overall, it outlines that Pou3f1 mediates the role of Nfatc3 in regulating macrophage infammation and carcinogenesis in UC-CRC development.
Keywords Ulcerative colitis-associated colorectal cancer, Macrophage, Pou3f1, Nfatc3, Infammation
Address and Contact Information 1 Department of Gastroenterology, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, 110004 Shenyang, China
2 Department of Gastroenterology, The Second Afliated Hospital of Shenyang Medical College, Shenyang, China
3 Department of Respiratory, Ansteel Group General Hospital, Anshan, China
4 Medical Oncology Ward, Tieling Central Hospital, Tieling, China
*Corresponing author: Lianjie Lin leanlj13@yeah.net
Read full article at BMC

No.  76DOI: 10.1186/s11658-022-00380-2 Volume 27 (2022) - 27:76
Title IDENTIFCATION OF COX4I2 AS A HYPOXIA‐ASSOCIATED GENE ACTING THROUGH FGF1 TO PROMOTE EMT AND ANGIOGENESIS IN CRC
Authors Jie‐pin Li1,2,3†, Yuan‐jie Liu2,3†, Shu‐hong Zeng2,3, Hai‐jian Gao1, Yu‐gen Chen2* and Xi Zou2,3,4*
Abstract Background: Current evidence suggests that the hypoxic tumor microenvironment further aggravates tumor progression, leading to poor therapeutic outcomes. There is as yet no biomarker capable of evaluating the hypoxic state of the tumor. The cytochrome c oxidase (COX) subunit is crucial to the mitochondrial respiratory chain. Methods: We investigated the potential oncogenic role of COX subunit 4 isoform 2 gene (COX4I2) in colorectal cancer (CRC) by least absolute shrinkage and selection operator (LASSO) and COX regression analysis to examine whether COX4I2 overexpression can predict colorectal cancer (CRC) prognosis. The association of COX4I2 levels with clinical features and its biological actions were evaluated both in vitro and in vivo. Results: Our analysis showed that elevated COX4I2 levels were correlated with poor clinical outcomes. We also observed that that COX4I2 may be involved in epithelial-mesenchymal transition, activation of cancer-related fbroblasts and angiogenesis in relation to fbroblast growth factor 1. Conclusions: The COX4I2 level may be a predictor of outcome in CRC and may represent a novel target for treatment development.
Keywords Colorectal cancer, COX4I2, Fibroblast growth factor 1, Epithelial–mesenchymal transition, Angiogenesis, Cancer-associated fbroblasts
Address and Contact Information 1 Zhangjiagang TCM Hospital Afliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu, China
2 Afliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, Jiangsu, China
3 No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
4 Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China
*Corresponding author: chenyg666@126.com; zxvery@126.com
Jie-pin Li and Yuan-jie Liu contributed equally to this work
Read full article at BMC

No.  77DOI: 10.1186/s11658-022-00382-0 Volume 27 (2022) - 27:77
Title DIVERSIFCATION OF PAR SIGNALING THROUGH RECEPTOR CROSSTALK
Authors Irene Lee‐Rivera, Edith López and Ana María López‐Colomé*
Abstract Protease activated receptors (PARs) are among the frst receptors shown to transactivate other receptors: noticeably, these interactions are not limited to members of the same family, but involve receptors as diverse as receptor kinases, prostanoid receptors, purinergic receptors and ionic channels among others. In this review, we will focus on the evidence for PAR interactions with members of their own family, as well as with other types of receptors. We will discuss recent evidence as well as what we consider as emerging areas to explore; from the signalling pathways triggered, to the physiological and pathological relevance of these interactions, since this additional level of molecular cross-talk between receptors and signaling pathways is only beginning to be explored and represents a novel mechanism providing diversity to receptor function and play important roles in physiology and disease.
Keywords Receptor dimerization, GPCR transactivation, Receptor cofactoring, Receptor signalling crosstalk
Address and Contact Information Department of Molecular Neuropathology, Instituto de Fisiología Celular, UNAM, Apartado Postal 70‐253, Ciudad Universitaria, Mexico City, CdMx, Mexico
*Corresponding author: acolome@ifc.unam.mx
Read full article at BMC

No.  78DOI: 10.1186/s11658-022-00387-9 Volume 27 (2022) - 27:78
Title CORRECTION: LABORATORY METHODS TO DECIPHER EPIGENETIC SIGNATURES: A COMPARATIVE REVIEW
Authors Raheleh Halabian1, Valizadeh Arshad2, Ali Ahmadi3, Pardis Saeedi1, Sadegh Azimzadeh Jamalkandi4 and Mohammad Reza Alivand5*
Abstract Correction: Cellular & Molecular Biology Letters (2021) 26:46 https://doi.org/10.1186/ s11658-021-00290-9.
Following publication of the original article [1], we have been informed that the afliation of the author Raheleh Halabian has been incorrectly assigned.
Te afliation group has been updated above and the original article has been corrected.

Published online 21 September 2022

References
1. Halabian, et al. Cellular & Molecular Biology Letters (2021) 26:46 https://doi.org/10.1186/s11658-021-00290-9.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional afliations.
Keywords
Address and Contact Information 1 1Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
2 Department of Stem Cell and Developmental Biology, Cell Science Research Center, Royan Institute For Stem Cell Biology and Technology, ACECR, Tehran, Iran
3 3Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
4 Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Mollasadra Ave, 14359-16471 Tehran, Iran
5 Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
*Corresponding author: Mohammad Reza Alivand alivand@yahoo.com
The online version of the original article can be found athttps://doi.org/10.1186/s11658-021-00290-9
Read full article at BMC

No.  79DOI: 10.1186/s11658-022-00370-4 Volume 27 (2022) - 27:79
Title Hsa‐microRNA‐27b‐3p INHIBITS HEPATOCELLULAR CARCINOMA PROGRESSION BY INACTIVATING TRANSFORMING GROWTH FACTOR‐ACTIVATED KINASE‐BINDING PROTEIN 3/NUCLEAR FACTOR KAPPA B SIGNALLING
Authors Jingyuan Wen1,2†, Zhao Huang1,2†, Yi Wei1,2†, Lin Xue1,2, Yufei Wang1,2, Jingyu Liao1,2, Junnan Liang1,2, Xiaoping Chen1,2,3, Liang Chu1,2* and Bixiang Zhang1,2,3*
Abstract Background: MicroRNAs (miRNAs) play crucial roles in the development of hepatocellular carcinoma (HCC). Hsa-microRNA-27b-3p (hsa-miR-27b) is involved in the formation and progression of various cancers, but its role and clinical value in HCC remain unclear.
Methods: The expression of hsa-miR-27b in HCC was examined by quantitative real-time PCR (qRT-PCR) and in situ hybridization (ISH) assays of clinical samples. Cell Counting Kit-8 assays (CCK-8), 5-ethynyl-2′-deoxyuridine (EdU) incorporation assays, Transwell assays, flamentous actin (F-actin) staining and western blot analyses were used to determine the efects of hsa-miR-27b on HCC cells in vitro. Subcutaneous xenograft and lung metastatic animal experiments were conducted to verify the role of hsa-miR-27b in HCC in vivo. In silico prediction, qRT-PCR, western blot, anti-Argonaute 2 (AGO2) RNA immunoprecipitation (RIP) and dual luciferase reporter assays were applied to identify the target genes of hsa-miR-27b. To detect the impacts of hsa-miR-27b on nuclear factor kappa B (NF-кB) signalling cascades mediated by transforming growth factor-activated kinase-binding protein 3 (TAB3), we performed qRT-PCR, western blot assays, immunofuorescence staining, immunohistochemistry (IHC) and dual-luciferase reporter assays. Recombinant oncolytic adenovirus (OncoAd) overexpressing hsa-miR-27b was constructed to detect their therapeutic value in HCC.
Results: The expression of hsa-miR-27b was lower in HCC than in adjacent non-tumourous tissues (ANTs), and the reduced expression of hsa-miR-27b was associated with worse outcomes in patients with HCC. hsa-miR-27b signifcantly inhibited the proliferation, migration, invasion, subcutaneous tumour growth and lung metastasis of HCC cells. The suppression of hsa-miR-27b promoted the nuclear translocation of NF-кB by upregulating TAB3 expression. TAB3 was highly expressed in HCC compared with ANTs and was negatively correlated with the expression of hsa-miR-27b. The impaired cell proliferation, migration and invasion by hsa-miR-27b overexpression were recovered by ectopic expression of TAB3. Recombinant OncoAd with overexpression of hsa-miR-27b induced anti-tumour activity compared with that induced by negative control (NC) OncoAd in vivo and in vitro.
Conclusions: By targeting TAB3, hsa-miR-27b acted as a tumour suppressor by inactivating the NF-кB pathway in HCC in vitro and in vivo, indicating its therapeutic value against HCC.
Keywords HCC, Hsa-miR-27b, NF-кB, TAB3, Oncolytic adenovirus
Address and Contact Information 1 Hepatic Surgery Center and Hubei Key Laboratory of Hepato-Biliary-Pancreatic Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
2 Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
3 Key Laboratory of Organ Transplantation, Ministry of Education; Key Laboratory of Organ Transplantation, National Health Commission; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Science, Wuhan, China
*Corresponding author: liangchu@tjh.tjmu.edu.cn; bixiangzhang@163.com
Jingyuan Wen, Zhao Huang and Yi Wei contributed equally to this work
Read full article at BMC

No.  80DOI: 10.1186/s11658-022-00372-2 Volume 27 (2022) - 27:80
Title circ_0086296 INDUCED ATHEROSCLEROTIC LESIONS VIA THE IFIT1/STAT1 FEEDBACK LOOP BY SPONGING miR‐576‐3p
Authors Min Zhang1*†, Yiqian Zhu2†, Jie Zhu3, Yi Xie1, Ruihao Wu1, JiaYin Zhong1, Zhaohui Qiu1* and Li Jiang1*
Abstract Extensive infammation of endothelial cells (ECs) facilitates atherosclerotic lesion formation. Circular RNA (circRNA) participates in atherosclerosis (AS)-related infammation responses; however, whether and how circ_0086296 regulates atherosclerotic infammation and lesions have not been investigated. Microarray analysis, quantitative real-time polymerase chain reaction, and fuorescence in situ hybridization assay were performed to detect the expression and location of hsa_circ_0086296 in human carotid artery plaques, aorta of atherosclerotic mice, and human umbilical vein endothelial cells (HUVECs). Sanger sequencing was used to verify the loop structure of circ_0086296. The relationship among circ_0086296, miR-576-3p, IFIT1, STAT1, and EIF4A3 was validated using bioinformatics, luciferase assay, RNA pull-down assay, and RNA immunoprecipitation. The atherosclerosis mouse model was used to evaluate the function of circ_0086296 in vivo. circ_0086296 expression was signifcantly upregulated in human carotid artery plaques, oxidized low-density lipoprotein (ox-LDL)-treated HUVECs, and the aorta of atherosclerotic mice. Functional analysis indicated that circ_0086296 promotes ECs injury in vitro and atherosclerosis progression in vivo. The mechanism analysis indicated that circ_0086296 sponged miR-576-3p to promote IFIT1–STAT1 expression. Moreover, STAT1 upregulated circ_0086296 expression, forming the circ_0086296/miR-576-3p/IFIT1/STAT1 feedback loop. Notably, inhibition of the circ_0086296/miR-576-3p/IFIT1 axis could block atherosclerotic lesion formation both in vivo and in vitro. Finally, circ_0086296 was overexpressed in exosomes of patients with atherosclerosis and exosomes of ox-LDL-treated ECs. Therefore, the circ_0086296/miR-576-3p/IFIT1/STAT1 feedback loop participates in atherosclerosis progression and contributes to the high circ_0086296 expression observed in the exosomes of serum of patients with atherosclerosis. This study sought to provide a deep understanding of the mechanisms underlying the aberrant EC phenotype in AS.
Keywords Atherosclerosis, circ_0086296, miR-576-3p, IFIT1, STAT1, Exosomes, EIF4A3
Address and Contact Information 1 Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
2 Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
3 Center for Translational Neurodegeneration and Regenerative Therapy, Tenth People’s Hospital of Tongji University, Shanghai, China
*Corresponding author: zm19821982@hotmail.com; ZhaohuiQiu@163.com; lijiang@sohu.com
Min Zhang and Yiqian Zhu are equally contributed to this paper.
Read full article at BMC

No.  81DOI: 10.1186/s11658-022-00383-z Volume 27 (2022) - 27:81
Title INHIBITION OF CISD2 PROMOTES FERROPTOSIS THROUGH FERRITINOPHAGY‐MEDIATED FERRITIN TURNOVER AND REGULATION OF p62–Keap1–NRF2 PATHWAY
Authors Yanchun Li1,2†, Bing Xu3†, Xueying Ren4†, Luyang Wang2, Yaqing Xu2, Yefeng Zhao2, Chen Yang2, Chen Yuan2, Huanjuan Li2, Xiangmin Tong2*, Ying Wang1* and Jing Du2*
Abstract Background: CDGSH iron sulfur domain 2 (CISD2) is an iron–sulfur protein with a [2Fe–2S] cluster, which is critical for cell proliferation and iron homeostasis. It has been demonstrated that aberrant expression of CISD2 is associated with the progression of multiple cancers. However, the underlying mechanism of CISD2 in regulating tumorigenesis remains obscure.
Methods: Bioinformatics strategies were used to investigate the protein interaction network and functional annotation of CISD2. In the functional experiment, cell viability was measured by CCK-8 kit. The levels of cellular reactive oxygen species (ROS), intra-cellular free iron, lipid peroxides, and lysosomal activity were determined by DCF-DA, RPA, C11-BODIPY, and cathepsin B staining, respectively. The glutathione (GSH) content was determined using a GSH assay kit.
Results: We showed that knockdown of CISD2 signifcantly accelerated the Erastin-induced ferroptotic cell death with excess lipid peroxidation, GSH exhaustion, and iron accumulation, while overexpression of CISD2 hindered the sensitivity to Erastin. Further assays via confocal microscopy and western blot exhibited that CISD2 knockdown markedly enhanced the lysosomal activity, and activated ferritinophagy under the exposure of Erastin. Pharmacological inhibition of lysosomal function could inhibit the degradation of ferritin heavy chain (FTH), and attenuate the phenotypes of ferroptosis, such as accelerated iron accumulation and lipid peroxidation. Notably, we found that Erastin-induced compensatory elevation of nuclear factor erythroid 2-related factor 2 (NRF2) could be eliminated in CISD2 depletion cells. Mechanically, CISD2 knockdown promoted the degradation of autophagy adaptor p62 and resulted in an increased binding afnity of Keap1 with NRF2, thus leading to the increased ubiquitination and subsequent degradation of NRF2. Enforced expression of NRF2 reversed the sensitivity of shCISD2 cells to ferroptosis both in vitro and in vivo. Conversely, enforced expression of Keap1 exacerbated the degradation of NRF2, reduced the transcriptional expression of FTH and heme oxygenase 1 (HO-1), increased the oxidative damage, and thus further facilitated ferroptosis.
Conclusion: Taken together, our current results illustrated two parallel mechanisms involved in the shCISD2-mediated ferroptosis. One was that shCISD2 enhanced the accumulation of free iron via ferritinophagy-dependent ferritin turnover; the other was that CISD2 depletion induced the inhibition of the p62–Keap1–NRF2 pathway, which resulted in oxidative stress and ferroptosis.
Keywords CISD2, Iron, Autophagy, Ferroptosis, Ferritinophagy
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 Afliated Hospital of Zhejiang Chinese Medical University, 310005 Hangzhou, 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
Read full article at BMC

No.  82DOI: 10.1186/s11658-022-00381-1 Volume 27 (2022) - 27:82
Title 5‐METHOXYFAVONE‐INDUCED AMPKα ACTIVATION INHIBITS NF‐κB AND P38 MAPK SIGNALING TO ATTENUATE INFUENZA A VIRUS‐MEDIATED INFAMMATION AND LUNG INJURY IN VITRO AND IN VIVO
Authors Sushan Yang1†, Linxin Wang2†, Xiping Pan2†, Yueyun Liang1, Yuehan Zhang1, Jing Li3,4* and Beixian Zhou1*
Abstract Infuenza-related acute lung injury (ALI) is a life-threatening condition that results mostly from uncontrolled replication of infuenza virus (IV) and severe proinfammatory responses. The methoxy favonoid compound 5-methoxyfavone (5-MF) is believed to have superior biological activity in the treatment of cancer. However, the efects and underlying mechanism of 5-MF on IV-mediated ALI are still unclear. Here, we showed that 5-MF signifcantly improved the survival of mice with lethal IV infection and ameliorated IV-mediated lung edema, lung histological changes, and infammatory cell lung recruitment. We found that 5-MF has antiviral activity against infuenza A virus (IAV), which was probably associated with increased expression of radical S-adenosyl methionine domain containing 2 (RSAD2) and suppression of endosomal acidifcation. Moreover, IV-infected A549 cells with 5-MF treatment markedly reduced proinfammatory mediator expression (IL-6, CXCL8, TNF-α, CXCL10, CCL2, CCL3, CCL4, GM-CSF, COX-2, and PGE2) and prevented P-IKBα, P-P65, and P-P38 activation. Interestingly, we demonstrated that 5-MF treatment could trigger activation of AMP-activated protein kinase (AMPK)α in IV-infected A549 cells, as evidenced by activation of the AMPKα downstream molecule P53. Importantly, the addition of AMPKα blocker compound C dramatically abolished 5-MF-mediated increased levels of RSAD2, the inhibitory efects on H1N1 virus-elicited endosomal acidifcation, and the suppression expression of proinfammatory mediators (IL-6, TNF-α, CXCL10, COX-2 and PGE2), as well as the inactivation of P-IKBα, P-P65, and P-P38 MAPK signaling pathways. Furthermore, inhibition of AMPKα abrogated the protective efects of 5-MF on H1N1 virus-mediated lung injury and excessive infammation in vivo. Taken together, these results indicate that 5-MF alleviated IV-mediated ALI and suppressed excessive infammatory responses through activation of AMPKα signaling.
Keywords 5-Methoxyfavone, Infuenza A virus, AMPKα, Anti-infammatory, Antiviral
Address and Contact Information 1 The People’s Hospital of Gaozhou, Gaozhou 525200, China
2 Guangzhou Laboratory, Guangzhou, China
3 State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Afliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
4 Institute of Chinese Integrative Medicine, Guangzhou Medical University, Guangzhou, Guangdong, China
*Correspondence: lijinghenan@163.com; zbeixian@126.com
Sushan Yang, Linxin Wang, and Xiping Pan contributed equally to this work.
Read full article at BMC

No.  83DOI: 10.1186/s11658-022-00384-y Volume 27 (2022) - 27:83
Title THE ROLE OF MACROPHAGE SUBTYPES AND EXOSOMES IN IMMUNOMODULATION
Authors Abdulwahab Tefischi Gharavi1†, Niloofar Asadi Hanjani1†, Elaheh Movahed2 and Mohammad Doroudian1*
Abstract Macrophages are infuential members of the innate immune system that can be reversibly polarized by diferent microenvironment signals. Cell polarization leads to a wide range of features, involving the migration, development, and organization of the cells. There is mounting evidence that macrophage polarization plays a key role in the initiation and development of a wide range of diseases. This study aims to give an overview of macrophage polarization, their diferent subtypes, and the importance of alternatively activated M2 macrophage and classically activated M1 macrophage in immune responses and pathological conditions. This review provides insight on the role of exosomes in M1/M2-like macrophage polarization and their potential as a promising therapeutic candidate.
Keywords Exosomes, Infammation, Macrophages, Macrophage polarization
Address and Contact Information 1 Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran 14911-15719, Iran
2 Wadsworth Center, New York State Department of Health, Albany, New Year, USA
*Corresponding author: mdoroudi@tcd.ie
Abdulwahab Tefischi Gharavi and Niloofar Asadi Hanjani contributed equally to this work.
Read full article at BMC

No.  84DOI: 10.1186/s11658-022-00385-x Volume 27 (2022) - 27:84
Title lncRNA PVT1: A NOVEL ONCOGENE IN MULTIPLE CANCERS
Authors Ruiming Li1, Xia Wang1, Chunming Zhu2* and Kefeng Wang1*
Abstract Long noncoding RNAs are involved in epigenetic gene modifcation, including binding to the chromatin rearrangement complex in pre-transcriptional regulation and to gene promoters in gene expression regulation, as well as acting as microRNA sponges to control messenger RNA levels in post-transcriptional regulation. An increasing number of studies have found that long noncoding RNA plasmacytoma variant translocation 1 (PVT1) plays an important role in cancer development. In this review of a large number of studies on PVT1, we found that PVT1 is closely related to tumor onset, proliferation, invasion, epithelial–mesenchymal transformation, and apoptosis, as well as poor prognosis and radiotherapy and chemotherapy resistance in some cancers. This review comprehensively describes PVT1 expression in various cancers and presents novel approaches to the diagnosis and treatment of cancer.
Keywords Long noncoding RNA, PVT1, Oncogene, Cancer
Address and Contact Information 1 Department of Urology, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
2 Department of Family Medicine, Shengjing Hospital of China Medical University, #36 Sanhao Street, Heping District, Shenyang 110004, Liaoning, China
*Corresponding author: chunmzhu@126.com; wang.kefeng@hotmail.com
Read full article at BMC

No.  85DOI: 10.1186/s11658-022-00379-9 Volume 27 (2022) - 27:85
Title THERAPEUTIC OVEREXPRESSION OF miR‐92a‐2‐5p AMELIORATED CARDIOMYOCYTE OXIDATIVE STRESS INJURY IN THE DEVELOPMENT OF DIABETIC CARDIOMYOPATHY
Authors Manli Yu1†, Yangyong Sun2†, Xinghua Shan1†, Fan Yang2, Guojun Chu1, Qian Chen2, Lin Han2*, Zhifu Guo1* and Guokun Wang2*
Abstract Background: Diabetic cardiomyopathy (DCM) results from pathological changes in cardiac structure and function caused by diabetes. Excessive oxidative stress is an important feature of DCM pathogenesis. MicroRNAs (miRNAs) are key regulators of oxidative stress in the cardiovascular system. In the present study, we screened for the expression of oxidative stress-responsive miRNAs in the development of DCM. Furthermore, we aimed to explore the mechanism and therapeutic potential of miR-92a-2-5p in preventing diabetes-induced myocardial damage.
Methods: An experimental type 2 diabetic (T2DM) rat model was induced using a high-fat diet and low-dose streptozotocin (30 mg/kg). Oxidative stress injury in cardiomyocytes was induced by high glucose (33 mmol/L). Oxidative stress-responsive miR-NAs were screened by quantitative real-time PCR. Intervention with miR-92a-2-5p was accomplished by tail vein injection of agomiR in vivo or adenovirus transfection in vitro.
Results: The expression of miR-92a-2-5p in the heart tissues was signifcantly decreased in the T2DM group. Decreased miR-92a-2-5p expression was also detected in high glucose-stimulated cardiomyocytes. Overexpression of miR-92a-2-5p attenuated cardiomyocyte oxidative stress injury, as demonstrated by increased glutathione level, and reduced reactive oxygen species accumulation, malondialdehyde and apoptosis levels. MAPK interacting serine/threonine kinase 2 (MKNK2) was verifed as a novel target of miR-92a-2-5p. Overexpression of miR-92a-2-5p in cardiomyocytes signifcantly inhibited MKNK2 expression, leading to decreased phosphorylation of p38-MAPK signaling, which, in turn, ameliorated cardiomyocyte oxidative stress injury. Additionally, diabetes-induced myocardial damage was signifcantly alleviated by the injection of miR-92a-2-5p agomiR, which manifested as a signifcant improvement in myocardial remodeling and function.
Conclusions: miR-92a-2-5p plays an important role in cardiac oxidative stress, and may serve as a therapeutic target in DCM.
Keywords Diabetic cardiomyopathy, Cardiomyocytes, Oxidative stress, MicroRNA, MAP kinase interacting serine/threonine kinase 2
Address and Contact Information 1 Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
2 Department of Cardiovascular Surgery, Institute of Cardiac Surgery, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
*Corresponding author: sh_hanlin@163.com; guozhifu@126.com; dearwgk@smmu.com.cn
Manli Yu, Yangyong Sun and Xinghua Shan contributed equally to this work
Read full article at BMC

No.  86DOI: 10.1186/s11658-022-00389-7 Volume 27 (2022) - 27:86
Title ANTI‐AGING EFECT OF β‐CAROTENE THROUGH REGULATING THE KAT7‐P15 SIGNALING AXIS, INFAMMATION AND OXIDATIVE STRESS PROCESS
Authors Wei V. Zheng1, Wang Xu4, Yaqin Li3, Jie Qin5, Tao Zhou1, Dezhi Li1, Yanwei Xu1, Xianyi Cheng1,2, Yu Xiong1,2 and Zaizhong Chen1,2*
Abstract Background: Research on aging is growing as the elderly make up a greater share of the population, focusing on reversing and inhibiting the aging process. The exhaustion and senescence of stem cells are the fundamental drivers behind aging. β-Carotene has been depicted to have many biological functions, and we speculate that it may have an anti-aging efect.
Methods: Firstly, the anti-aging property of β-carotene was investigated in vitro using mesenchymal stem cells (MSCs) induced by H2O2. The anti-aging efect was characterized using Western-bloting, confocal laser scanning microscopy, indirect immunofuorescence, and immunohistochemistry. The anti-aging property was also tested in vivo using aged mice.
Results: The in vitro experiment revealed that β-carotene could relieve the aging of MSCs, as evidenced by a series of aging marker molecules such as p16 and p21. β-Carotene appeared to inhibit aging by regulating the KAT7-P15 signaling axis. The in vivo experiment revealed that β-carotene treatment has signifcantly down-regulated the aging level of tissues and organs.
Conclusions: In this work, we explored the anti-aging efect of β-carotene in vivo and in vitro. The experimental results indicate that β-carotene may be an important potential anti-aging molecule, which can be used as a drug or in functional food to treat aging in the future.
Keywords β-Carotene, Aging, Stem cells, KAT7-P15 signaling axis
Address and Contact Information 1 Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People’s Republic of China
2 Department of Minimally Invasion Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People’s Republic of China
3 Department of Infectious Disease, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People’s Republic of China
4 College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
5 Scientifc and Resaerch Dept., Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, People’s Republic of China
*Corresponding author: chenzaizhong2021@yeah.net
Read full article at BMC

No.  87DOI: 10.1186/s11658-022-00390-0 Volume 27 (2022) - 27:87
Title EXOSOMAL DNAJB11 PROMOTES THE DEVELOPMENT OF PANCREATIC CANCER BY MODULATING THE EGFR/MAPK PATHWAY
Authors Peng Liu1,2, Fuqiang Zu1,2, Hui Chen1, Xiaoli Yin3*† and Xiaodong Tan1,2*†
Abstract Pancreatic ductal adenocarcinoma (PDAC) is a malignant tumor with invasive and metastatic characteristics and poor prognosis. Intracellular protein homeostasis is associated with invasion and metastasis of pancreatic cancer, but the specifc molecular mechanism remains unclear. Our previous studies have revealed that DNAJB11, a key protein in protein homeostasis, is secreted by exosomes in the supernatant of dissociated pancreatic cancer cells with high metastasis. The results from transcriptome sequencing and co-immunoprecipitation (Co-IP)-based liquid chromatography with tandem mass spectrometry (LC–MS/MS) showed that depletion of DNAJB11 levels could increase HSPA5 expression and induce endoplasmic reticulum stress through the PRKR-like endoplasmic reticulum kinase signaling pathway in pancreatic cancer cells. Furthermore, exosomal DNAJB11 promoted cell development of PC cells in vitro and in vivo. In addition, exosomal DNAJB11 could regulate the expression of EGFR and activate the downstream MAPK signaling pathway. Clinical blood samples were collected to evaluate the potential of exosome DNAJB11 as a diagnostic biomarker and therapeutic target for the treatment of pancreatic cancer. This study could provide a new theoretical basis and potential molecular targets for the treatment of pancreatic cancer.
Keywords Pancreatic cancer, Exosomal protein, Signal transduction, Early diagnosis
Address and Contact Information 1 Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, China
2 Diagnostic and Therapeutic Center of Pancreatic Diseases of Liaoning Province, Shenyang 110004, China
3 Department of Radiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
*Corresponding author: yinxiaoli057728@163.com; tanxdcmu@163.com
Xiaoli Yin and Xiaodong Tan contributed equally to this work
Read full article at BMC

No.  88DOI: 10.1186/s11658-022-00393-x Volume 27 (2022) - 27:88
Title DOWNREGULATION OF EXOSOMAL miR‐7‐5p PROMOTES BREAST CANCER MIGRATION AND INVASION BY TARGETING RYK AND PARTICIPATING IN THE ATYPICAL WNT SIGNALLING PATHWAY
Authors Zhaoyi Liang, Lu Liu, Ruixia Gao, Chengchuan Che and Ge Yang*
Abstract Background: Current studies show that exosomal miRNAs become an important factor in cancer metastasis. Among the many miRNA studies, miR-7-5p has not been thoroughly investigated in breast cancer metastasis.
Methods: Bioinformatic screening was performed using extant data from the GEO database, and miR-7-5p expression levels in breast cancer cell lines and exosomes were further examined using real-time quantitative PCR (qRT-PCR). The extracted exosomes were characterised by transmission electron microscopy (TEM), particle size analysis and marker protein determination. Cell migration and invasion were then examined using wound healing assays and Transwell assays, respectively. Correlation between miR-7-5p and receptor-like tyrosine kinase (RYK) was analysed by luciferase reporter. The efect of miR-7-5p against RYK-related downstream factors was verifed using western blot assays.
Results: In this study, we found that the expression of miR-7-5p was signifcantly diferent in exosomes secreted from breast cancer cell lines with diferent high and low invasiveness. Further experiments revealed that miR-7-5p has an important role in inhibiting the migration and invasion of breast cancer. In terms of mechanism of action, miR-7-5p was found to target the RYK, leading to its reduced expression, which in turn caused a reduction in the phosphorylation level of the downstream factor JNK. Reduced levels of phosphorylated JNK factors lead to reduced levels of phosphorylation of c-Jun protein, which in turn leads to increased expression of EMT transcription factors, thereby inhibiting the epithelial–mesenchymal transition (EMT) process to suppress the invasion of breast cancer.
Conclusion: Thus, we demonstrated that exosomes loaded with high levels of miR-7-5p emitted from less aggressive breast cancers can participate in the atypical WNT pathway by targeting the RYK gene and thus inhibit breast cancer metastasis.
Keywords Breast cancer, Exosome, miR-7-5p, RYK, EMT, Atypical WNT pathway
Address and Contact Information College of Life Sciences, Qufu Normal University, Qufu 273165, Shandong, China
*Corresponding author: yangge@qfnu.edu.cn
Read full article at BMC

No.  89DOI: 10.1186/s11658-022-00386-w Volume 27 (2022) - 27:89
Title LncRNA SNHG16 PROMOTES DEVELOPMENT OF OESOPHAGEAL SQUAMOUS CELL CARCINOMA BY INTERACTING WITH EIF4A3 AND MODULATING RhoU mRNA STABILITY
Authors Lihua Ren1, Xin Fang1, Sachin Mulmi Shrestha1, Qinghua Ji1, Hui Ye1, Yan Liang1, Yang Liu1, Yadong Feng1, Jingwu Dong2 and Ruihua Shi1*
Abstract Background: Numerous studies have revealed that long noncoding RNAs (lncRNAs) are closely related to the development of many diseases and carcinogenesis. However, their specifc biological function and molecular mechanism in oesophageal squamous cell carcinoma (ESCC) remains unclear.
Methods: RNA-Seq was performed to determine the diferential expressions of lncR-NAs in ESCC, and the level of SNHG16 expression was detected in ESCC and intraepithelial neoplasia (IEN) samples. In vitro and in vivo experiments were performed to explore the role of SNHG16 and the interaction of EIF4A3 and Ras homologue family member U (RhoU) signalling.
Results: One hundred and seventy-fve upregulated and 134 downregulated lncRNAs were identifed by RNA-Seq. SNHG16 was highly expressed in ESCC and intraepithelial neoplasia (IEN) samples, and its expression level was correlated with tumour diferentiation and T stage. Overexpression of SNHG16 can facilitate ESCC cell proliferation and metastasis. Mechanistically, we noticed that SNHG16 could bind RNA binding protein (RBP)-eukaryotic translation initiation factor (EIF4A3) and interact with it to form a complex. Importantly, the coalition of SNHG16 and EIF4A3 ultimately regulated Ras homologue family member U (RhoU). SNHG16 modulated RhoU expression by recruiting EIF4A3 to regulate the stability of RhoU mRNA. Knockdown of RhoU further alleviated the efect of the SNHG16 oncogene in ESCC cells.
Conclusions: The newly identifed SNHG16–EIF4A3–RhoU signalling pathway directly coordinates the response in ESCC pathogenesis and suggests that SNHG16 is a promising target for potential ESCC treatment.
Keywords SNHG16, Oesophageal squamous cell carcinoma, mRNA stability, EIF4A3, RhoU
Address and Contact Information 1 Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing 210009, Jiangsu Province, People’s Republic of China
2 Department of Gastroenterology, Xuyi County People’s Hospital, Huaian 211700, People’s Republic of China *Corresponding author: ruihuashi@126.com
Read full article at BMC

No.  90DOI: 10.1186/s11658-022-00392-y Volume 27 (2022) - 27:90
Title E2F1 PROMOTES CELL CYCLE PROGRESSION BY STABILIZING SPINDLE FIBER IN COLORECTAL CANCER CELLS
Authors Zejun Fang1,2,3†, Min Lin1†, Shenghui Chen3,4, Hong Liu5, Minjing Zhu2, Yanyan Hu2, Shanshan Han2, Yizhang Wang2, Long Sun6, Fengjiao Zhu2*, Chengfu Xu3* and Chaoju Gong7*
Abstract Background: E2F1 is a transcription factor that regulates cell cycle progression. It is highly expressed in most cancer cells and activates transcription of cell cycle-related kinases. Stathmin1 and transforming acidic coiled-coil-containing protein 3 (TACC3) are factors that enhance the stability of spindle fiber.
Methods: The E2F1-mediated transcription of transforming acidic coiled-coil-con-taining protein 3 (TACC3) and stathmin1 was examined using the Cancer Genome Atlas (TCGA) analysis, quantitative polymerase chain reaction (qPCR), immunoblotting, chromatin immunoprecipitation (ChIP), and luciferase reporter. Protein–protein interaction was studied using co-IP. The spindle structure was shown by immunofuorescence. Phenotype experiments were performed through MTS assay, fow cytometry, and tumor xenografts. Clinical colorectal cancer (CRC) specimens were analyzed based on immunohistochemistry.
Results: The present study showed that E2F1 expression correlates positively with the expression levels of stathmin1 and TACC3 in colorectal cancer (CRC) tissues, and that E2F1 transactivates stathmin1 and TACC3 in CRC cells. Furthermore, protein kinase A (PKA)-mediated phosphorylation of stathmin1 at Ser16 is essential to the phosphorylation of TACC3 at Ser558, facilitating the assembly of TACC3/clathrin/α-tubulin complexes during spindle formation. Overexpression of Ser16-mutated stathmin1, as well as knockdown of stathmin1 or TACC3, lead to ectopic spindle poles including disorganized and multipolar spindles. Overexpression of wild-type but not Ser16-mutated stathmin1 promotes cell proliferation in vitro and tumor growth in vivo. Consistently, a high level of E2F1, stathmin1, or TACC3 not only associates with tumor size, lymph node metastasis, TNM stage, and distant metastasis, but predicts poor survival in CRC patients.
Conclusions: E2F1 drives the cell cycle of CRC by promoting spindle assembly, in which E2F1-induced stathmin1 and TACC3 enhance the stability of spindle fiber.
Keywords E2F1, Stathmin1, TACC3, Colorectal carcinoma, Spindle fber, Cell cycle
Address and Contact Information 1 Central Laboratory, Sanmen People’s Hospital of Zhejiang Province, Sanmen 317100, China
2 Department of Clinical Laboratory, Sanmen People’s Hospital of Zhejiang Province, No. 15 Taihe Road, Hairun Street, Sanmen 317100, China
3 Department of Gastroenterology, The First Afliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou 310003, China
4 Department of Gastroenterology, Afliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
5 Department of Microbiology, Immunology and Infammation, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
6 Department of Gastrointestinal Surgery, Sanmen People’s Hospital of Zhejiang Province, Sanmen 317100, China
7 Central Laboratory, The Afliated Xuzhou Municipal Hospital of Xuzhou Medical University, No. 19 Zhongshan Bei Road, Xuzhou 221100, China
*Corresponding author: sM.zFj@126.com; xiaofu@zju.edu.cn; gongcj@zju.edu.cn
Zejun Fang and Min Lin contributed equally to this work
Read full article at BMC

No.  91DOI: 10.1186/s11658-022-00376-y Volume 27 (2022) - 27:91
Title LARRPM RESTRICTS LUNG ADENOCARCINOMA PROGRESSION AND M2 MACROPHAGE POLARIZATION THROUGH EPIGENETICALLY REGULATING LINC00240 AND CSF1
Authors Yue Li, Chen Chen, Hai‐lin Liu, Zhen‐fa Zhang and Chang‐li Wang*
Abstract Background: Long non-coding RNAs (lncRNAs) are critical regulators in lung adeno‐carcinoma (LUAD). M2-type tumor-associated macrophages (TAMs) also play oncogenic roles in LUAD. However, the involvement of lncRNAs in TAM activation is still largely unknown.
Methods: The expressions of LARRPM, LINC00240 and CSF1 were determined by RT-qPCR. The regulation of LINC00240 and CSF1 by LARRPM was investigated by RNA–protein pull-down, RNA immunoprecipitation, chromatin immunoprecipitation and bisulfte DNA sequencing. In vitro and in vivo gain- and loss-of-function assays were performed to investigate the roles of LARRPM.
Results: The lncRNA LARRPM was expressed at low levels in LUAD tissues and cells. The low expression of LARRPM was correlated with advanced stage and poor survival of patients with LUAD. Functional experiments revealed that LARRPM suppressed LUAD cell proliferation, migration and invasion, and promoted apoptosis. LARRPM also repressed macrophage M2 polarization and infltration. Taken together, LARRPM signifcantly restricted LUAD progression in vivo. Mechanistically, LARRPM bound and recruited DNA demethylase TET1 to the promoter of its anti-sense strand gene LINC00240, leading to a decrease in DNA methylation level of the LINC00240 promoter and transcriptional activation of LINC00240. Functional rescue assays suggested that the lncRNA LINC00240 was responsible for the roles of LARRPM in the malignant behavior of LUAD cells. LARRPM decreased the binding of TET1 to the CSF1 promoter, resulting in increased DNA methylation of the CSF1 promoter and transcriptional repression of CSF1, which is responsible for the roles of LARRPM in macrophage M2 polarization and infltration. The TAMs educated by LUAD cells exerted oncogenic roles, which was negatively regulated by LARRPM expressed in LUAD cells.
Conclusions: LARRPM restricts LUAD progression through repressing both LUAD cell and macrophages. These data shed new insights into the regulation of LUAD progression by lncRNAs and provide data on the potential utility of LARRPM as a target for LUAD treatment.
Keywords Tumor-associated macrophage, Lung adenocarcinoma, Progression, DNA methylation, CSF1
Address and Contact Information Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin 300060, China
*Corresponding author: changli_w@163.com
Read full article at BMC

No.  92DOI: 10.1186/s11658-022-00394-w Volume 27 (2022) - 27:92
Title TARGETING UBR5 IN HEPATOCELLULAR CARCINOMA CELLS AND PRECISE TREATMENT VIA ECHINACOSIDE NANODELIVERY
Authors Menghan Wang1†, Xing Ma1†, Guoyu Wang1, Yanan Song2, Miao Zhang2, Zhongchao Mai1, Borong Zhou1, Ying Ye2* and Wei Xia1*
Abstract Background: Hepatocellular carcinoma (HCC) is among the most common and malignant cancers with no efective therapeutic approaches. Echinacoside (ECH), a phenylethanoid glycoside isolated from Chinese herbal medicine, Cistanche salsa, can inhibit HCC progression; however, poor absorption and low bioavailability limit its biological applications.
Methods: To improve ECH sensitivity to HepG2 cells, we developed a mesoporous silica nanoparticle (MSN)-based drug delivery system to deliver ECH to HepG2 cells via galactose (GAL) and poly(ethylene glycol) diglycidyl ether (PEGDE) conjugation (ECH@ Au@MSN-PEGDE-GAL, or ECH@AMPG). Gain- and loss-of-function assays were conducted to assess the efects of UBR5 on HCC cell apoptosis and glycolysis. Moreover, the interactions among intermediate products were also investigated to elucidate the mechanisms by which UBR5 functions.
Results: The present study showed that ubiquitin protein ligase E3 component N-recognin 5 (UBR5) acted as an oncogene in HCC tissues and that its expression was inhibited by ECH. AMPG showed a high drug loading property and a slow and sustained release pattern over time. Moreover, owing to the valid drug accumulation, ECH@AMPG promoted apoptosis and inhibited glycolysis of HepG2 cells in vitro. In vivo experiments demonstrated that AMPG also enhanced the antitumor efects of ECH in HepG2 cell-bearing mice.
Conclusions: Our results indicated the clinical signifcance of UBR5 as a therapeutic target. On the basis of the nontoxic and high drug-loading capabilities of AMPG, ECH@ AMPG presented better efects on HCC cells compared with free ECH, indicating its potential for the chemotherapy of HCC.
Keywords Nanoparticle, Echinacoside, Drug delivery, Hepatocellular carcinoma, UBR5
Address and Contact Information 1 Department of Nuclear Medicine, The Seventh People’s Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai 200137, China 2 Central Laboratory, The Seventh People’s Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Rd, Pudong New Area, Shanghai 200137, China
*Corresponding author: yy49453324@163.com; tcm_xiawei@163.com
Menghan Wang and Xing Ma contributed equally to this work
Read full article at BMC

No.  93DOI: 10.1186/s11658-022-00388-8 Volume 27 (2022) - 27:93
Title DOWN‐REGULATION OF EVA1A BY miR‐103a‐3p PROMOTES HEPATOCELLULAR CARCINOMA CELLS PROLIFERATION AND MIGRATION
Authors Qian Xu1†, Zhaozhong Liao1†, Zunshuang Gong1†, Xiaokun Liu1, Yuling Yang2, Zhe Wang3, Weiyan Yang4, lin Hou1, Jiejie Yang1, Junying Song1, Wenjing Liu1, Bin Wang5, Junnan Hua3, Mingyi Pu2 and Ning Li1*
Abstract Background: EVA1A (Eva-1 homolog A), a novel protein involved in autophagy and apoptosis, functions as a tumor suppressor in some human primary cancers, including hepatocellular carcinoma (HCC). While it is consistently downregulated in several cancers, its involvement in hepatocarcinogenesis is still largely unknown.
Methods: We frst detected the expression of EVA1A in HCC tissues and cell lines using RT‒qPCR, immunohistochemistry and western blotting and detected the expression of miR-103a-3p by RT‒qPCR. Then, bioinformatics prediction, dual-luciferase reporter gene assays and western blotting were used to screen and identify the upstream microRNA of EVA1A. After manipulating the expression of miR-103a-3p or EVA1A, wound healing, invasion, proliferation, colony formation, apoptosis, autophagy, mitosis and mitochondrial function assays, including mitochondrial membrane potential, ROS and ATP production assays, were performed to investigate the functions of miR-103a-3p targeting EVA1A in HCC cells. Apoptosis-related proteins were assessed by RT‒qPCR (TP53) or western blotting (TP53, BAX, Bcl-2 and caspase-3). Autophagy level was evaluated by observing LC3 puncta and examining the protein levels of p62, Beclin1 and LC3-II/I.
Results: We found that EVA1A expression was decreased while miR-103a-3p expression was increased in HCC tissues and cell lines and that their expression was inversely correlated in HCC patients. The expression of miR-103a-3p was associated with HCC tumor stage and poor prognosis. miR-103a-3p could target EVA1A through direct binding to its 3’-UTR and suppress its expression. Overexpression of miR-103a-3p signifcantly downregulated the expression of EVA1A, TP53 and BAX, upregulated the JAK2/STAT3 pathway and promoted HCC cell migration, invasion and proliferation, while repression of miR-103a-3p dramatically upregulated the expression of EVA1A, TP53, BAX and cleaved-caspase-3, inhibited HCC cell migration, invasion and proliferation, and caused mitochondrial dysfunction and apoptosis. Overexpression of EVA1A signifcantly attenuated the cancer-promoting efects of miR-103a-3p in HCC cells, while knockdown of EVA1A alleviated the mitochondrial dysfunction and apoptosis caused by miR-103a-3p inhibition. Overexpression of EVA1A did not induce signifcant changes in autophagy levels, nor did it afect G2/M transition or mitosis.
Conclusion: These fndings indicate that the downregulation of the tumor suppressor EVA1A by miR-103a-3p potentially acts as a key mediator in HCC progression, mainly by inhibiting apoptosis and promoting metastasis. The miR-103a/EVA1A/TP53 axis provides a new potential diagnostic and therapeutic target for HCC treatment.
Keywords EVA1A, miR-103a-3p, hepatocellular carcinoma, Apoptosis, TP53, Autophagy, Mitosis
Address and Contact Information 1 Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, Qingdao, China
2 Department of Infectious Diseases, Afliated Hospital of Qingdao University, Qingdao, China
3 Department of Biotechnology, School of Basic Medicine, Qingdao University, Qingdao, China
4 Department of Anesthesiology, Family Planning Service Center, Maternal and Child Health Hospital of Jiaozhou City, Qingdao, China
5 College of Electronic Information, Micro-Nano Technology College, Qingdao University, Qingdao, China
*Corresponding author: ning-99@163.com
Qian Xu, Zhaozhong Liao and Zunshuang Gong contributed equally to this work.
Read full article at BMC

No.  94DOI: 10.1186/s11658-022-00364-2 Volume 27 (2022) - 27:94
Title circ_0025033 PROMOTES OVARIAN CANCER DEVELOPMENT VIA REGULATING THE hsa_miR‐370‐3p/SLC1A5 AXIS
Authors Huiping Ma1, Shuyun Qu2, Yao Zhai2 and Xiaofeng Yang1*
Abstract Background: Circular RNAs (circRNAs) appear to be important modulators in ovarian cancer. We aimed to explore the role and mechanism of circ_0025033 in ovarian cancer.
Methods: qRT-PCR was conducted to determine circ_0025033, hsa_miR-370-3p, and SLC1A5 mRNA expression. Functional experiments were conducted, including Cell Counting Kit-8 (CCK-8), 5-ethynyl-2′-deoxyuridine (EdU), fow cytometry, transwell, tube formation, xenograft tumor model assay, western blot analysis of protein levels, and analysis of glutamine metabolism using commercial kits. Their predicted interaction was confrmed using dual-luciferase reporter and RNA pull-down.
Results: circ_0025033 was upregulated in ovarian cancer; its knockdown induced proliferation, invasion, angiogenesis, glutamine metabolism, and apoptosis in vitro, and blocked tumor growth in vivo. circ_0025033 regulated ovarian cancer cellular behaviors via sponging hsa_miR-370-3p. In parallel, SLC1A5 might abolish the anti-ovarian cancer role of hsa_miR-370-3p. Furthermore, circ_0025033 afected SLC1A5 via regulating hsa_miR-370-3p.
Conclusion: circ_0025033 might promote ovarian cancer progression via hsa_miR-370-3p/SLC1A5, providing an interesting insight into ovarian cancer tumorigenesis.

Highlights
• circ_0025033 knockdown inhibited ovarian cancer malignant behaviors.
• circ_0025033 served as a hsa_miR-370-3p sponge.
• hsa_miR-370-3p targeted SLC1A5.
Keywords Ovarian cancer, circ_0025033, hsa_miR-370-3p, SLC1A5
Address and Contact Information 1 Department of Gynecology, First Afliated Hospital of Xi’an Jiaotong University, No. 277 Yanta West Road, Xi’an 710000, Shaanxi, China
2 Department of Gynecology, Gynaecology Hospital of Shaanxi Nuclear Industry, Xi’an, Shaanxi, China
*Corresponding author: yangxf73@126.com
Read full article at BMC

No.  95DOI: 10.1186/s11658-022-00395-9 Volume 27 (2022) - 27:95
Title CIRCULATING SMALL EXTRACELLULAR VESICLE‐ENCAPSULATED SEMA5A‐IT1 ATTENUATES MYOCARDIAL ISCHEMIA–REPERFUSION INJURY AFTER CARDIAC SURGERY WITH CARDIOPULMONARY BYPASS
Authors Ting Wu1,2, Guoning Shi2, Zhenhua Ji2, Shu Wang2, Lizhu Geng2 and Zhigang Guo3*
Abstract Cardiomyocyte injury is a common complication during cardiac surgery with cardiopulmonary bypass (CPB). Studies have shown that circulating small extracellular vesicles (sEVs) are involved in the pathological process of cardiovascular diseases via delivering signaling molecules. This study aims to investigate the relationship between circulating sEV-encapsulated long noncoding RNAs (lncRNAs) and cardiac injury after CPB. Here, we found that the expression of sEV SEMA5A-IT1 in serum samples of patients after CPB was higher than that of pre-CPB serum samples. Moreover, serum-derived sEV SEMA5A-IT1 levels were negatively correlated with creatine kinase-MB (CK-MB) levels in patients who underwent CPB operation. Notably, circulating sEVs packaged with SEMA5A-IT1 could be uptaken by cardiomyocyte-like cells AC16 and increased SEMA5A-IT1 expression in AC16 cells. Upregulated SEMA5A-IT1 protected cardiomyocytes against hypoxia/reoxygenation injury, confrmed by increased cell viability, reduced cell apoptosis, and inhibited ferroptosis in AC16 cells. Mechanistically, SEMA5A-IT1 regulated the expression of B-cell CLL/lymphoma 2 (BCL2) and solute carrier family 7 member 11 (SLC7A11) through sponging miR-143-3p. Transfection of miR-143-3p mimics, BCL2, or SLC7A11 knockdown could attenuate the protective efect of SEMA5A-IT1 on cardiomyocytes. In conclusion, we propose that SEMA5A-IT1, which is transported to cardiomyocytes through circulating sEVs, is an important regulatory molecule that protects cardiomyocytes from ischemia–reperfusion injury, providing a target for the prevention and treatment of myocardial ischemia–reperfusion injury.
Keywords Cardiopulmonary bypass, Cardiomyocyte injury, Small extracellular vesicles, microRNA, Apoptosis, Ferroptosis
Address and Contact Information 1 Department of Cardiopulmonary Bypass, Clinical College of Chest, Tianjin Medical University, Tianjin, China
2 Department of Cardiopulmonary Bypass, Chest Hospital, Tianjin University, Tianjin, China
3 Department of Cardiovascular Surgery, Chest Hospital, Tianjin University, No. 261, Taierzhuang South Road, Jinnan, Tianjin 300222, China
*Corresponding author: gzgang1118@163.com
Read full article at BMC

No.  96DOI: 10.1186/s11658-022-00396-8 Volume 27 (2022) - 27:96
Title CORRECTION: GRANULIN AS AN IMPORTANT IMMUNE MOLECULE INVOLVED IN LAMPREY TISSUE REPAIR AND REGENERATION BY PROMOTING CELL PROLIFERATION AND MIGRATION
Authors Ruixiang Sun1,2,3†, Dong Wang1,2,3†, Yuxuan Song1,2,3†, Qingwei Li1,2,3, Peng Su1,2,3* and Yue Pang1,2,3*
Abstract Correction: Cellular & Molecular Biology Letters (2022) 27:64
https://doi.org/10.1186/s11658-022-00360-6
Following publication of the original article [1], the authors identifed an error in Fig. 5. They reused the picture of NC group of Fig. 5D (published on line) in the PGRN-S1 group. Thus, they used the correct picture to replace it. The incorrect and the correct figure is given below.
Te incorrect Fig. 5 is available online at https://cmbl.biomedcentral.com/articles/10.1186/s11658-022-00396-8
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional afliations.
Keywords
Address and Contact Information 1 College of Life Sciences, Liaoning Normal University, Dalian 116081, China
2 Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
3 Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
*Corresponding author: sp4046@163.com; pangyue01@163.com
Ruixiang Sun, Dong Wang and Yuxuan Song contributed equally to this work
The original article can be found online at https://doi.org/10.1186/s11658-022-00360-6.
Read full article at BMC

No.  97DOI: https://cmbl.biomedcentral.com/articles/10.1186/s11658-022-00399-510.1186/s11658-022-00399-5 Volume 27 (2022) - 27:97
Title X‐box BINDING PROTEIN 1 AS A KEY MODULATOR IN “HEALING ENDOTHELIAL CELLS”, A NOVEL EC PHENOTYPE PROMOTING ANGIOGENESIS AFTER MCAO
Authors Zhuohui Chen1,2,4, Xiang Wang1,2, Haiyue Wu1,2, Yishu Fan1,2, Zhouyi Yan1,2, Chenxiao Lu1,2, Hongfei Ouyang1,2, Shiyu Zhang1,2 and Mengqi Zhang1,3*
Abstract Background: Endothelial cells (ECs) play an important role in angiogenesis and vascular reconstruction in the pathophysiology of ischemic stroke. Previous investigations have provided a profound cerebral vascular atlas under physiological conditions, but have failed to identify new disease-related cell subtypes. We aimed to identify new EC subtypes and determine the key modulator genes.
Methods: Two datasets GSE174574 and GSE137482 were included in the study. Seurat was utilized as the standard quality-control pipeline. UCell was used to calculate singlecell scores to validate cellular identity. Monocle3 and CytoTRACE were utilized in aid of pseudo-time diferentiation analysis. CellChat was utilized to infer the intercellular communication pathways. The angiogenesis ability of ECs was validated by MTS, Transwell, tube formation, fow cytometry, and immunofuorescence assays in vitro and in vivo. A synchrotron radiation-based propagation contrast imaging was introduced to comprehensively portray cerebral vasculature.
Results: We successfully identifed a novel subtype of EC named “healing EC” that highly expressed pan-EC marker and pro-angiogenic genes but lowly expressed all the arteriovenous markers identifed in the vascular single-cell atlas. Further analyses showed its high stemness to diferentiate into other EC subtypes and potential to modulate infammation and angiogenesis via excretion of signal molecules. We therefore identifed X-box binding protein 1 (Xbp1) as a key modulator in the healing EC phenotype. In vitro and in vivo experiments confrmed its pro-angiogenic roles under both physiological and pathological conditions. Synchrotron radiation-based propagation contrast imaging further proved that Xbp1 could promote angiogenesis and recover normal vasculature conformation, especially in the corpus striatum and prefrontal cortex under middle cerebral artery occlusion (MCAO) condition.
Conclusions: Our study identifed a novel disease-related EC subtype that showed high stemness to diferentiate into other EC subtypes. The predicted molecule Xbp1 was thus confrmed as a key modulator that can promote angiogenesis and recover normal vasculature conformation.
Keywords Single-cell RNA sequencing, Endothelial cell, Middle cerebral artery occlusion, X-box binding protein 1, Synchrotron radiation
Address and Contact Information 1 Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, China
2 Xiangya School of Medicine, Central South University, Changsha 410013, China
3 National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
4 Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, China
*Corresponding author: zhangmengqi8912@163.com
Read full article at BMC

No.  98DOI: 10.1186/s11658-022-00391-z Volume 27 (2022) - 27:98
Title FAT STORAGE‐INDUCING TRANSMEMBRANE PROTEINS: BEYOND MEDIATING LIPID DROPLET FORMATION
Authors Gaiping Wang*, Anqi Chen, Yu Wu, Danlin Wang, Cuifang Chang and Guoying Yu*
Abstract Fat storage-inducing transmembrane proteins (FITMs) were initially identifed in 2007 as members of a conserved endoplasmic reticulum (ER) resident transmembrane protein gene family, and were found to be involved in lipid droplet (LD) formation. Recently, several studies have further demonstrated that the ability of FITMs to directly bind to triglyceride and diacylglycerol, and the diphosphatase activity of hydrolyzing fatty acyl-CoA, might enable FITMs to maintain the formation of lipid droplets, engage in lipid metabolism, and protect against cellular stress. Based on the distribution of FITMs in tissues and their important roles in lipid droplet biology and lipid metabolism, it was discovered that FITMs were closely related to muscle development, adipocyte diferentiation, and energy metabolism. Accordingly, the abnormal expression of FITMs was not only associated with type 2 diabetes and lipodystrophy, but also with cardiac disease and several types of cancer. This study reviews the structure, distribution, expression regulation, and functionality of FITMs and their potential relationships with various metabolic diseases, hoping to provide inspiration for fruitful research directions and applications of FITM proteins. Moreover, this review will provide an important theoretical basis for the application of FITMs in the diagnosis and treatment of related diseases.
Keywords Fat storage-inducing transmembrane proteins (FITMs), Lipid droplets (LDs), Lipid metabolism, Endoplasmic reticulum stress, Metabolic disease
Address and Contact Information State Key Laboratory Cell Diferentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, Henan, China
*Corresponding author: xiaowang0529@163.com; 2018207@htu.edu.cn
Read full article at BMC

No.  99DOI: 10.1186/s11658-022-00398-6 Volume 27 (2022) - 27:99
Title tRF‐3013b INHIBITS GALLBLADDER CANCER PROLIFERATION BY TARGETING TPRG1L
Authors Lu Zou1,2,3†, Yang Yang1,2,3†, Biyu Zhou4†, Weijian Li1,2,3, Ke Liu1,2,3, Guoqiang Li1,2,3, Huijie Miao1,2,3, Xiaoling Song2,5, Jiahua Yang1,2,3, Yajun Geng1,2,3, Maolan Li1,2,3*, Runfa Bao2,5* and Yingbin Liu1,2,3*
Abstract Background: tRNA-derived fragments (tRFs) are newly discovered noncoding RNAs and regulate tumor progression via diverse molecular mechanisms. However, the expression and biofunction of tRFs in gallbladder cancer (GBC) have not been reported yet.
Methods: The expression of tRFs in GBC was detected by tRF and tiRNA sequencing in GBC tissues and adjacent tissues. The biological function of tRFs was investigated by cell proliferation assay, clonal formation assay, cell cycle assay, and xenotransplantation model in GBC cell lines. The molecular mechanism was discovered and verifed by transcriptome sequencing, fuorescence in situ hybridization (FISH), target gene site prediction, and RNA binding protein immunoprecipitation (RIP).
Results: tRF-3013b was signifcantly downregulated in GBC compared with paracancer tissues. Decreased expression of tRF-3013b in GBC patients was correlated with poor overall survival. Dicer regulated the production of tRF-3013b, and its expression was positively correlated with tRF-3013b in GBC tissues. Functional experiments demonstrated that tRF-3013b inhibited GBC cell proliferation and induced cell-cycle arrest. Mechanically, tRF-3013b exerted RNA silencing efect on TPRG1L by binding to AGO3, and then inhibited NF-κB. TPRG1L overexpression could rescue the efects of tRF-3013b on GBC cell proliferation.
Conclusions: This study indicated that Dicer-induced tRF-3013b inhibited GBC proliferation by targeting TPRG1L and repressed NF-κB, pointing to tRF-3013b as a novel potential therapeutic target of GBC.
Keywords Gallbladder cancer, tRNA-derived fragments, Cell cycle, Cell proliferation
Address and Contact Information 1 Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
2 Shanghai Key Laboratory of Biliary Tract Disease Research, Shanghai 200092, China
3 Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
4 Department of Plastic and Reconstructive Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China
5 Department of General Surgery, Xinhua Hospital Afliated to Shanghai Jiao Tong University School of Medicine, 1665, Kongjiang Road, Shanghai 200092, China
*Corresponding author: limaolan6@163.com; baorunfa@yeah.net; laoniulyb@shsmu.edu.cn
Lu Zou, Yang Yang, and Biyu Zhou contributed equally to this work
Read full article at BMC

No.  100DOI: 10.1186/s11658-022-00400-1 Volume 27 (2022) - 27:100
Title ACQUIRED DRUG RESISTANCE INTERFERES WITH THE SUSCEPTIBILITY OF PROSTATE CANCER CELLS TO METABOLIC STRESS
Authors Jessica Catapano1, Marcin Luty1, Tomasz Wróbel1, Maciej Pudełek1, Katarzyna Piwowarczyk1, Sylwia Kędracka‐Krok2,4, Maciej Siedlar3, Zbigniew Madeja1 and Jarosław Czyż1*
Abstract Background: Metformin is an inhibitor of oxidative phosphorylation that displays an array of anticancer activities. The interference of metformin with the activity of multidrug resistance systems in cancer cells has been reported. However, the consequences of the acquired chemoresistance for the adaptative responses of cancer cells to metformin-induced stress and for their phenotypic evolution remain unaddressed.
Methods: Using a range of phenotypic and metabolic assays, we assessed the sensitivity of human prostate cancer PC-3 and DU145 cells, and their drug-resistant lineages (PC-3_DCX20 and DU145_DCX20), to combined docetaxel/metformin stress. Their adaptation responses have been assessed, in particular the shifts in their metabolic profle and invasiveness.
Results: Metformin increased the sensitivity of PC-3 wild-type (WT) cells to docetaxel, as illustrated by the attenuation of their motility, proliferation, and viability after the combined drug application. These efects correlated with the accumulation of energy carriers (NAD(P)H and ATP) and with the inactivation of ABC drug transporters in docetaxel/metformin-treated PC-3 WT cells. Both PC-3 WT and PC-3_DCX20 reacted to metformin with the Warburg efect; however, PC-3_DCX20 cells were considerably less susceptible to the cytostatic/misbalancing efects of metformin. Concomitantly, an epithelial–mesenchymal transition and Cx43 upregulation was seen in these cells, but not in other more docetaxel/metformin-sensitive DU145_DCX20 populations. Stronger cytostatic efects of the combined fenofbrate/docetaxel treatment confrmed that the fne-tuning of the balance between energy supply and expenditure determines cellular welfare under metabolic stress.
Conclusions: Collectively, our data identify the mechanisms that underlie the limited potential of metformin for the chemotherapy of drug-resistant tumors. Metformin can enhance the sensitivity of cancer cells to chemotherapy by inducing their metabolic decoupling/imbalance. However, the acquired chemoresistance of cancer cells impairs this efect, facilitates cellular adaptation to metabolic stress, and prompts the invasive front formation.
Keywords Metabolic stress, Drug resistance, Prostate cancer, Metabolic adaptation, Metformin
Address and Contact Information 1 Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30‐387 Kraków, Poland
2 Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30‐387 Kraków, Poland
3 Department of Clinical Immunology, Institute of Pediatrics, Faculty of Medicine, Jagiellonian University Medical College, Wielicka 265, 30‐663 Kraków, Poland
4 Proteomics and Mass Spectrometry Laboratory, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, 30‐387 Kraków, Poland
*Corresponding author: jarek.czyz@uj.edu.pl
Read full article at BMC

No.  101DOI: 10.1186/s11658-022-00404-x Volume 27 (2022) - 27:101
Title ALKBH5 IN MOUSE TESTICULAR SERTOLI CELLS REGULATES Cdh2 mRNA TRANSLATION TO MAINTAIN BLOOD–TESTIS BARRIER INTEGRITY
Authors Zhonglin Cai1,2,3, Yao Zhang2, Lin Yang2, Chunhui Ma2, Yi Fei2, Jing Ding2, Wei Song4, Wei‐Min Tong2,5*, Yamei Niu2,5* and Hongjun Li1*
Abstract Background: RNA N6-methyladenosine (m6A) is involved in mammalian spermatogenesis. In both germ cells and Leydig cells, ALKBH5 regulates spermatogenesis and androgen synthesis in an m6A-dependent manner. However, it is unclear whether ALKBH5 plays a role in testicular Sertoli cells, which constitute the blood–testis barrier (BTB) through cell junctions between adjacent Sertoli cells. Methods: ALKBH5 expression in the testes of humans and mice was detected by immunohistochemical staining and immunofuorescence staining. BTB integrity was evaluated by BTB assay. m6A-seq was performed to screen for BTB-related molecules regulated by ALKBH5. m6A immunoprecipitation–quantitative real-time polymerase chain reaction (qPCR), RNA immunoprecipitation–qPCR, western blot, coimmunoprecipitation, and polysome fractionation–qPCR analyses were performed to explore the mechanisms of ALKBH5 in BTB. Transmission electron microscopy was applied to observe the BTB ultrastructure. Results: ALKBH5 in Sertoli cells is related to the integrity of the BTB. Subsequently, the m6A level on Cdh2 mRNA, encoding a structural protein N-cadherin in the BTB, was found to be regulated by ALKBH5. IGF2BP1/2/3 complexes and YTHDF1 promoted Cdh2 mRNA translation. In addition, we found that basal endoplasmic specialization, in which N-cadherin is a main structural protein, was severely disordered in the testes of Alkbh5-knockout mice. Conclusions: Our study revealed that ALKBH5 regulates BTB integrity via basal endoplasmic specialization by afecting Cdh2 mRNA translation.
Keywords RNA N6-methyladenosine, Alkbh5, Blood–testis barrier, Cdh2, Basal endoplasmic specialization
Address and Contact Information 1 Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
2 Department of Pathology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
3 Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
4 Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
5 Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
*Corresponding author: wmtong@ibms.pumc.edu. cn; niuym@ibms.pumc.edu.cn; lihongjun@pumch.cn
Read full article at BMC

No.  102DOI: 10.1186/s11658-022-00401-0 Volume 27 (2022) - 27:102
Title A NOVEL SERUM SPHERICAL LECTIN FROM LAMPREY REVEALS A MORE EFCIENT MECHANISM OF IMMUNE INITIATION AND REGULATION IN JAWLESS VERTEBRATES
Authors Jiali Lu1,2†, Jinsong Duan3†, Yinglun Han1,2†, Meng Gou1,2, Jun Li1,2, Qingwei Li1,2 and Yue Pang1,2*
Abstract The innate immune system is the body’s frst line of defense against pathogens and involves antibody and complement system-mediated antigen removal. Immune-response-related complement molecules have been identifed in lamprey, and the occurrence of innate immune response via the mannose-binding lectin-associated serine proteases of the lectin cascade has been reported. We have previously shown that lamprey (Lampetra japonica) serum can efciently and specifcally eliminate foreign pathogens. Therefore, we aimed to understand the immune mechanism of lamprey serum in this study. We identifed and purifed a novel spherical lectin (LSSL) from lamprey serum. LSSL had two structural calcium ions coordinated with conserved amino acids, as determined through cryogenic electron microscopy. LSSL showed high binding capacity with microbial and mammalian glycans and demonstrated agglutination activity against bacteria. Phylogenetic analysis revealed that LSSL was transferred from phage transposons to the lamprey genome via horizontal gene transfer. Furthermore, LSSL was associated with mannose-binding lectin-associated serine protease 1 and promoted the deposition of the C3 fragment on the surface of target cells upon binding. These results led us to conclude that LSSL initiates and regulates agglutination, resulting in exogenous pathogen and tumor cell eradication. Our observations will give a greater understanding of the origin and evolution of the complement system in higher vertebrates and lead to the identifcation of novel immune molecules and pathways for defense against pathogens and tumor cells.

Key points

• A novel serum spherical lectin from lamprey was identifed which have agglutination activity against bacteria dependent on calcium.
• Te crystal structure of LSSL was determined, and adhered to both microbial and mammalian glycans with high binding capacity.
• Te role of LSSL was associated with MASP-1 and recruited C3 deposition for pathogen elimination.
Keywords Innate immunity, Lamprey, LSSL, Serum
Address and Contact Information 1 College of Life Sciences, Liaoning Normal University, Dalian 116081, China
2 Lamprey Research Center, Liaoning Normal University, Dalian 116081, China
3 State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
*Corresponding author: pangyue@lnnu.edu.cn
Jiali Lu, Jinsong Duan, and Yinglun Han contributed equally to this work
Read full article at BMC

No.  103DOI: 10.1186/s11658-022-00397-7 Volume 27 (2022) - 27:103
Title INSIGHT INTO THE PHYSIOLOGICAL AND PATHOLOGICAL ROLES OF THE ARYL HYDROCARBON RECEPTOR PATHWAY IN GLUCOSE HOMEOSTASIS, INSULIN RESISTANCE, AND DIABETES DEVELOPMENT
Authors Tahseen S. Sayed1, Zaid H. Maayah1, Heba A. Zeidan2, Abdelali Agouni1 and Hesham M. Korashy1*
Abstract The aryl hydrocarbon receptor (AhR) is a ligand-activated transcriptional factor that mediates the toxicities of several environmental pollutants. Decades of research have been carried out to understand the role of AhR as a novel mechanism for disease development. Its involvement in the pathogenesis of cancer, cardiovascular diseases, rheumatoid arthritis, and systemic lupus erythematosus have long been known. One of the current hot research topics is investigating the role of AhR activation by environmental pollutants on glucose homeostasis and insulin secretion, and hence the pathogenesis of diabetes mellitus. To date, epidemiological studies have suggested that persistent exposure to environmental contaminants such as dioxins, with subsequent AhR activation increases the risk of specifc comorbidities such as obesity and diabetes. The importance of AhR signaling in various molecular pathways highlights that the role of this receptor is far beyond just xenobiotic metabolism. The present review aims at providing signifcant insight into the physiological and pathological role of AhR and its regulated enzymes, such as cytochrome P450 1A1 (CYP1A1) and CYP1B1 in both types of diabetes. It also provides a comprehensive summary of the current fndings of recent research studies investigating the role of the AhR/CYP1A1 pathway in insulin secretion and glucose hemostasis in the pancreas, liver, and adipose tissues. This review further highlights the molecular mechanisms involved, such as gluconeogenesis, hypoxia-inducible factor (HIF), oxidative stress, and nfammation.
Keywords Aryl hydrocarbon receptor, Diabetes mellitus, Glucose hemostasis, Insulin, Environmental toxicants
Address and Contact Information 1 Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, 2713, Doha, Qatar
2 American School of Doha, Doha, Qatar
*Corresponding author: hkorashy@qu.edu.qa
Read full article at BMC

No.  104DOI: 10.1186/s11658-022-00405-w Volume 27 (2022) - 27:104
Title A RARE MUTATION (p.F149del) OF THE NT5C3A GENE IS ASSOCIATED WITH PYRIMIDINE 5′‐NUCLEOTIDASE DEFCIENCY
Authors Dżamila M. Bogusławska1*, Michał Skulski2, Rafał Bartoszewski3, Beata Machnicka1, Elżbieta Heger1, Kazimierz Kuliczkowski4 and Aleksander F. Sikorski5
Abstract Pyrimidine 5′-nucleotidase defciency is a rare erythrocyte enzymopathy. Here we report two cases of hemolytic anemia in brothers of Polish origin that are associated with a very rare mutation. Heterozygous deletion in the NT5C3A gene (c.444_446delGTT), inherited most likely from their asymptomatic mother, resulted in a single amino acid residue deletion (p.F149del) in cytosolic pyrimidine 5′-nucleotidase. However, only the mutated transcript was present in the reticulocyte transcriptome of both patients. Only residual activity of pyrimidine 5′-nucleotidase in the brothers’ erythrocytes could be observed when compared with the controls, including their asymptomatic father and sister. Western blot showed no sign of the presence of 5′-nucleotidase protein in the erythrocytes of both studied patients. The 2.5-fold reduction of the purine/pyrimidine ratio observed only in the brothers’ erythrocytes confrms the correlation of the results of molecular analysis, including whole-exome sequencing, with the phenotype of the pyrimidine 5′-nucleotidase defciency. Altogether, our results may substantiate the hypothesis of the heterogeneity of the molecular basis of the defect involving both the mutation presented here and negative regulation of expression of the “normal” allele.
Keywords Pyrimidine 5′-nucleotidase defciency, Hereditary hemolytic anemia, Erythrocyte enzymopathy, Pyrimidine metabolism, Whole-exome sequencing
Address and Contact Information 1 Department of Biotechnology, Institute of Biological Sciences, University of Zielona Góra, Prof. Z. Szafrana 1 St., 65-516 Zielona Góra, Poland
2 Department of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a St., 50-383 Wrocław, Poland
3 Department of Biophysics, Faculty of Biotechnology, University of Wrocław, F. Joliot-Curie 14a St., 50-383 Wrocław, Poland
4 Silesian Park of Medical Technology Kardio-Med Silesia, M. Curie-Skłodowskiej 10C St., 41-800 Zabrze, Poland
5 Research and Development Centre, Regional Specialist Hospital, Kamieńskiego 73a St., 51-154 Wrocław, Poland
*Corresponding author: d.boguslawska@wnb.uz.zgora.pl
Read full article at BMC

No.  105DOI: 10.1186/s11658-022-00403-y Volume 27 (2022) - 27:105
Title TARGETING URIDINE–CYTIDINE KINASE 2 INDUCED CELL CYCLE ARREST THROUGH DUAL MECHANISM AND COULD IMPROVE THE IMMUNE RESPONSE OF HEPATOCELLULAR CARCINOMA
Authors Dehai Wu1†, Congyi Zhang1†, Guanqun Liao2†, Kaiming Leng3†, Bowen Dong4†, Yang Yu1, Huilin Tai5, Lining Huang6, Feng Luo1, Bin Zhang1, Tiexiang Zhan7, Qiuhui Hu8 and Sheng Tai1*
Abstract Background: Pyrimidine metabolism is critical for tumour progression. Uridine–cytidine kinase 2 (UCK2), a key regulator of pyrimidine metabolism, is elevated during hepatocellular carcinoma (HCC) development and exhibits carcinogenic efects. However, the key mechanism of UCK2 promoting HCC and the therapeutic value of UCK2 are still undefned. The aim of this study is to investigate the potential of UCK2 as a therapeutic target for HCC.
Methods: Gene expression matrices were obtained from public databases. RNA-seq, co-immunoprecipitation and RNA-binding protein immunoprecipitation were used to determine the mechanism of UCK2 promoting HCC. Immune cell infltration level and immune-related functional scores were evaluated to assess the link between tumour microenvironment and UCK2.
Results: In HCC, the expression of UCK2 was upregulated in part by TGFβ1 stimulation. UCK2 promoted cell cycle progression of HCC by preventing the degradation of mTOR protein and maintaining the stability of PDPK1 mRNA. We also identifed UCK2 as a novel RNA-binding protein. Downregulation of UCK2 induced cell cycle arrest and activated the TNFα/NFκB signalling pathway-related senescence-associated secretory phenotype to modify the tumour microenvironment. Additionally, UCK2 was a bio-marker of the immunosuppressive microenvironment. Downregulated UCK2 induced a secretory phenotype, which could improve the microenvironment, and decreased UCK2 remodelling metabolism could lower the resistance of tumour cells to T-cell-mediated killing.
Conclusions: Targeting UCK2 inhibits HCC progression and could improve the response to immunotherapy in patients with HCC. Our study suggests that UCK2 could be an ideal target for HCC.
Keywords Uridine–cytidine kinase 2, Hepatocellular carcinoma, Microenvironment, Secretory phenotype, Pyrimidine metabolism
Address and Contact Information 1 Department of Hepatic Surgery, Second Afliated Hospital of Harbin Medical University, #246Xuefu Road, Harbin 150086, Heilongjiang, China
2 Department of Hepatobiliary Surgery, Foshan Hospital Afliated to Southern Medical University, Foshan 528000, China
3 Department of Hepatobiliary Surgery, Qingdao Municipal Hospital, Qingdao 266071, China
4 Department of Biochemistry & Molecular Biology, Harbin Medical University, Harbin 150081, China
5 McGill Mathematics and Statistics Department, Montreal, Canada
6 Department of Hepatobiliary Surgery, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215008, China
7 Department of Intensive Care Unit, Seventh Afliated Hospital of Sun Yat-Sen University, Shenzhen 528406, China
8 Department of Hepatobiliary Surgery, Second Cancer Hospital of Heilongjiang Province, Harbin 150088, China
*Corresponding author: Taisheng1973@163.com
Dehai Wu, Congyi Zhang, Guanqun Liao, Kaiming Leng and Bowen Dong contributed equally to this work
Read full article at BMC

No.  106DOI: 10.1186/s11658-022-00406-9 Volume 27 (2022) - 27:106
Title EXOSOMAL miR‐628‐5p FROM M1 POLARIZED MACROPHAGES HINDERS m6A MODIFCATION OF circFUT8 TO SUPPRESS HEPATOCELLULAR CARCINOMA PROGRESSION
Authors Liyan Wang, Xiaoyuan Yi, Xuhua Xiao, Qinghua Zheng, Lei Ma and Bin Li*
Abstract Background: Hepatocellular carcinoma (HCC) is the most common type of liver cancer. CircFUT8 has been shown to be upregulated in cancers, but its function in HCC remains unclear. Tumor-associated macrophages (TAMs) are one of the main components of the tumor microenvironment (TME), and M1 macrophages function as tumor suppressors in cancers. Exosomes exert an important role in the TME, and circRNAs can be modifed by m6A. We investigated the function of circFUT8 in HCC and its interaction with exosomes, M1 macrophages, and m6A.
Methods: CircFUT8 expression was detected in HCC cells, and its efects on HCC cell growth were verifed through functional assays. Mechanism assays including RNA pull down, RNA-binding protein immunoprecipitation (RIP), and luciferase reporter assays were undertaken to verify how circFUT8 may interact with miR-628-5p, and how these molecules may modulate HCC cell malignancy via interacting with exosomes and macrophages.
Results: CircFUT8 was upregulated in HCC cells and it accelerated HCC cell growth. Exosomes derived from M1 macrophages transferred miR-628-5p to HCC cells to inhibit human methyltransferase-like 14 (METTL14) expression. METTL14 promoted circFUT8 m6A modifcation and facilitated its nuclear export to the cytoplasm, where M1 macrophages regulated the circFUT8/miR-552-3p/CHMP4B pathway, thereby suppressing HCC progression.
Conclusion: M1 macrophages-derived exosomal miR-628-5p inhibited the m6A modifcation of circFUT8, inhibiting HCC development.
Keywords Hepatocellular carcinoma, Exosomes, M1 macrophages, circFUT8, m6A modifcation
Address and Contact Information Digestive Department, Afliated Hospital of Guilin Medical College, No.15 Lequn Road, Xiufeng District, Guilin 541001, Guangxi, China
*Corresponding author: Leebien@163.com
Read full article at BMC

No.  107DOI: /10.1186/s11658-022-00407-8 Volume 27 (2022) - 27:107
Title MF‐094 NANODELIVERY INHIBITS ORAL SQUAMOUS CELL CARCINOMA BY TARGETING USP30
Authors Xinyu Zhang1,2,3†, Yong Han1,2,3†, Shuli Liu1,2,3, Bing Guo4, Shengming Xu1,2,3, Yue He1,2,3* and Liu Liu1,2,3*
Abstract Background: Oral squamous cell carcinoma (OSCC) is a common head and neck cancer, and the incidence of OSCC is increasing. As the mortality of OSCC keeps increasing, it is crucial to clarify its pathogenesis and develop new therapeutic strategies.
Methods: Confocal laser scanning microscopy was used to evaluate the uptake of nanoparticles (NPs). The potential functions of USP30 were evaluated by cell counting kit (CCK)-8, fow cytometry, biochemical assay, coimmunoprecipitation, qRT–PCR, and immunoblotting. The antitumor efect of NP-loaded USP30 inhibitor MF-094 was evaluated in vitro and in vivo.
Results: In this study, increased USP30 expression was found in OSCC specimens and cell lines through qRT–PCR and immunoblotting. CCK-8, fow cytometry, and biochemical assay revealed that the deubiquitylated catalytic activity of USP30 contributed to cell viability and glutamine consumption of OSCC. Subsequently, USP30 inhibitor MF-094 was loaded in ZIF-8-PDA and PEGTK to fabricate ZIF-8-PDA-PEGTK nanoparticles, which exhibited excellent inhibition of cell viability and glutamine consumption of OSCC, both in vitro and in vivo.
Conclusion: The results indicated the clinical signifcance of USP30 and showed that nanocomposites provide a targeted drug delivery system for treating OSCC.
Keywords Head and neck cancer, Nanoparticles, ZIF-8, Ubiquitination, c-Myc
Address and Contact Information 1 Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, No.639 Zhizaoju Road, Huangpu District, Shanghai 200011, China
2 National Clinical Research Center for Oral Diseases, Shanghai, China
3 Shanghai Key Laboratory of Stomatology and Shanghai, Research Institute of Stomatology, Shanghai, China
4 Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
*Corresponding author: William5218@126.com; liuliu_618@163.com
Xinyu Zhang and Yong Han contributed equally to this work
Read full article at BMC

No.  108DOI: 10.1186/s11658-022-00402-z Volume 27 (2022) - 27:108
Title ABSENT IN MELANOMA 2 (AIM2) IN RHEUMATOID ARTHRITIS: NOVEL MOLECULAR INSIGHTS AND IMPLICATIONS
Authors Jianan Zhao1,2,6, Shicheng Guo3,4*, Steven J. Schrodi3,4* and Dongyi He1,2,5,6*
Abstract Absent in melanoma 2 (AIM2), a member of the Pyrin and HIN domain protein family, is a cytoplasmic receptor that recognizes double-stranded DNA. AIM2 exhibits limited expression under physiological conditions but is widely expressed in many human diseases, including autoimmune diseases, and plays an essential role in the immune response. Rheumatoid arthritis (RA) is an autoimmune disease that poses a severe threat to physical and mental health, and is caused by several genetic and metabolic factors. Multiple immune cells interact to form a complex infammatory network that mediates infammatory responses and bone destruction. Abnormal AIM2 expression in multiple immune cell populations (T cells, B cells, fbroblast-like synoviocytes, monocytes, and macrophages) may regulate multiple functional responses in RA through mechanisms such as pyroptosis, PANoptosis, and regulation of other molecules. In this review, we describe and summarize the functional regulation and impact of AIM2 expression in immune cells to improve our understanding of the complex pathological mechanisms. These insights may provide potential directions for the development of new clinical diagnostic strategies for RA.
Keywords Rheumatoid arthritis, Autoimmune disease, Absent in melanoma 2, Infammation, Pyroptosis, PANoptosis
Address and Contact Information 1 Department of Rheumatology, Shanghai Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
2 Guanghua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
3 Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI, USA
4 Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
5 Arthritis Institute of Integrated Traditional and Western Medicine, Shanghai Chinese Medicine Research Institute, Shanghai, China
6 Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
*Correspondence: Shicheng.Guo@wisc.edu; Schrodi@wisc.edu; dongyihe@medmail.com.cn
Read full article at BMC

No.  109DOI: 10.1186/s11658-022-00408-7 Volume 27 (2022) - 27:109
Title THE TRANSITION FROM HIF‐1 TO HIF‐2 DURING PROLONGED HYPOXIA RESULTS FROM REACTIVATION OF PHDs AND HIF1A mRNA INSTABILITY
Authors Maciej Jaśkiewicz1†, Adrianna Moszyńska2†, Jarosław Króliczewski2, Aleksandra Cabaj3, Sylwia Bartoszewska4, Agata Charzyńska3, Magda Gebert2, Michał Dąbrowski3, James F. Collawn5 and Rafal Bartoszewski6*
Abstract The hypoxia-inducible factors (HIF) are transcription factors that activate the adaptive hypoxic response when oxygen levels are low. The HIF transcriptional program increases oxygen delivery by inducing angiogenesis and by promoting metabolic reprograming that favors glycolysis. The two major HIFs, HIF-1 and HIF-2, mediate this response during prolonged hypoxia in an overlapping and sequential fashion that is referred to as the HIF switch. Both HIF proteins consist of an unstable alpha chain and a stable beta chain. The instability of the alpha chains is mediated by prolyl hydroxylase (PHD) activity during normoxic conditions, which leads to ubiquitination and proteasomal degradation of the alpha chains. During normoxic conditions, very little HIF-1 or HIF-2 alpha–beta dimers are present because of PHD activity. During hypoxia, however, PHD activity is suppressed, and HIF dimers are stable. Here we demonstrate that HIF-1 expression is maximal after 4 h of hypoxia in primary endothelial cells and then is dramatically reduced by 8 h. In contrast, HIF-2 is maximal at 8 h and remains elevated up to 24 h. There are diferences in the HIF-1 and HIF-2 transcriptional profles, and therefore understanding how the transition between them occurs is important and not clearly understood. Here we demonstrate that the HIF-1 to HIF-2 transition during prolonged hypoxia is mediated by two mechanisms: (1) the HIF-1 driven increase in the glycolytic pathways that reactivates PHD activity and (2) the much less stable mRNA levels of HIF-1α (HIF1A) compared to HIF-2α (EPAS1) mRNA. We also demonstrate that the alpha mRNA levels directly correlate to the relative alpha protein levels, and therefore to the more stable HIF-2 expression during prolonged hypoxia.
Keywords Hypoxia, Human endothelial cells, HIF1A, EPAS1, HIF-1α, HIF-2α
Address and Contact Information 1 International Research Agenda 3P- Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
2 Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
3 Laboratory of Bioinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
4 Department of Inorganic Chemistry, Medical University of Gdansk, Gdansk, Poland
5 Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, BirminghamBirmingham, AL 35233, USA
6 Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383 Wroclaw, Poland
*Corresponding author: rafal.bartoszewski@uwr.edu.pl
Maciej Jaśkiewicz and Adrianna Moszyńska contributed equally to this work
Read full article at BMC