1
|
Su Y, He S, Chen Q, Zhang H, Huang C, Zhao Q, Pu Y, He X, Jiang L, Ma Y, Zhao Q. Integrative ATAC-seq and RNA-seq analysis of myogenic differentiation of ovine skeletal muscle satellite cell. Genomics 2024; 116:110851. [PMID: 38692440 DOI: 10.1016/j.ygeno.2024.110851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/01/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
Skeletal muscle satellite cells (SMSCs) play an important role in regulating muscle growth and regeneration. Chromatin accessibility allows physical interactions that synergistically regulate gene expression through enhancers, promoters, insulators, and chromatin binding factors. However, the chromatin accessibility altas and its regulatory role in ovine myoblast differentiation is still unclear. Therefore, ATAC-seq and RNA-seq analysis were performed on ovine SMSCs at the proliferation stage (SCG) and differentiation stage (SCD). 17,460 DARs (differential accessibility regions) and 3732 DEGs (differentially expressed genes) were identified. Based on joint analysis of ATAC-seq and RNA-seq, we revealed that PI3K-Akt, TGF-β and other signaling pathways regulated SMSCs differentiation. We identified two novel candidate genes, FZD5 and MAP2K6, which may affect the proliferation and differentiation of SMSCs. Our data identify potential cis regulatory elements of ovine SMSCs. This study can provide a reference for exploring the mechanisms of the differentiation and regeneration of SMSCs in the future.
Collapse
Affiliation(s)
- Yingxiao Su
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China
| | - Siqi He
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China; College of Animal Science, Shanxi Agricultural University, Taigu 030801, China
| | - Qian Chen
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China; College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hechun Zhang
- Chaoyang Chaomu Breeding Farm Co., LTD, Chaoyang, Liaoning 122629, China
| | - Chang Huang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China; College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Qian Zhao
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China; College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yabin Pu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China
| | - Xiaohong He
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China
| | - Lin Jiang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China
| | - Yuehui Ma
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China
| | - Qianjun Zhao
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193,China.
| |
Collapse
|
2
|
Wu F, Zhang J, Jiang Q, Li Q, Li F, Li J, Lv W, Wang X, Qin Y, Huang C, Zhang S. MyoD1 promotes the transcription of BIK and plays an apoptosis-promoting role in the development of gastric cancer. Cell Cycle 2024; 23:573-587. [PMID: 38701194 PMCID: PMC11135814 DOI: 10.1080/15384101.2024.2348344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/23/2024] [Indexed: 05/05/2024] Open
Abstract
Myogenic differentiation (MyoD) 1, which is known as a pivotal transcription factor during myogenesis, has been proven dysregulated in several cancers. However, litter is known about the precise role and downstream genes of MyoD1 in gastric cancer (GC) cells. Here, we report that MyoD1 is lowly expressed in primary GC tissues and cells. In our experiments, overexpression of MyoD1 inhibited cell proliferation. Downstream genes of MyoD1 regulation were investigated using RNA-Seq. As a result, 138 up-regulated genes and 20 down-regulated genes and 27 up-regulated lncRNAs and 20 down-regulated lncRNAs were identified in MyoD1 overexpressed MKN-45 cells, which participated in epithelial cell signaling in Helicobacter pylori infection, glycosaminoglycan biosynthesis (keratan sulfate), notch signaling pathway, and others. Among these genes, BIK was directly regulated by MyoD1 in GC cells and inhibited cancer cell proliferation. The BIK knockdown rescued the effects of MyoD1 overexpression on GC cells. In conclusion, MyoD1 inhibited cell proliferation via 158 genes and 47 lncRNAs downstream directly or indirectly that participated in multiple signaling pathways in GC, and among these, MyoD1 promotes BIK transcription by binding to its promoter, then promotes BIK-Bcl2-caspase 3 axis and regulates GC cell apoptosis.
Collapse
Affiliation(s)
- Fei Wu
- Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Biomedical Experiment Center, Xian Jiaotong University, Xi’an, China
| | - Jinyuan Zhang
- Institute of Genetics and Development Biology, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, China
| | - Qiuyu Jiang
- Institute of Genetics and Development Biology, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, China
| | - Qian Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, China
| | - Fang Li
- Institute of Genetics and Development Biology, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, China
| | - Jia Li
- Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wei Lv
- Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaofei Wang
- Biomedical Experiment Center, Xian Jiaotong University, Xi’an, China
| | - Yannan Qin
- Institute of Genetics and Development Biology, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, China
| | - Chen Huang
- Institute of Genetics and Development Biology, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, China
| | - Shuqun Zhang
- Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
3
|
Militi S, Nibhani R, Jalali M, Pauklin S. RBL2-E2F-GCN5 guide cell fate decisions during tissue specification by regulating cell-cycle-dependent fluctuations of non-cell-autonomous signaling. Cell Rep 2023; 42:113146. [PMID: 37725511 DOI: 10.1016/j.celrep.2023.113146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/30/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023] Open
Abstract
The retinoblastoma family proteins (RBs) and E2F transcription factors are cell-autonomous regulators of cell-cycle progression, but they also impact fate choice in addition to tumor suppression. The range of mechanisms involved remains to be uncovered. Here, we show that RBs, particularly RBL2/p130, repress WNT ligands such as WNT4 and WNT8A, thereby directing ectoderm specification between neural crest to neuroepithelium. RBL2 achieves this function through cell-cycle-dependent cooperation with E2Fs and GCN5 on the regulatory regions of WNT loci, which direct neuroepithelial versus neural crest specification by temporal fluctuations of WNT/β-catenin and DLL/NOTCH signaling activity. Thus, the RB-E2F bona fide cell-autonomous axis controls cell fate decisions, and RBL2 regulates field effects via WNT ligands. This reveals a non-cell-autonomous function of RBL2-E2F in stem cell and tissue progenitor differentiation that has broader implications for cell-cycle-dependent cell fate specification in organogenesis, adult stem cells, tissue homeostasis, and tumorigenesis.
Collapse
Affiliation(s)
- Stefania Militi
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford OX3 7LD, UK
| | - Reshma Nibhani
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford OX3 7LD, UK
| | - Morteza Jalali
- Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Old Road, Headington, Oxford OX3 7LD, UK.
| |
Collapse
|
4
|
Mehboob R, Kurdi M, Ahmad M, Gilani SA, Khalid S, Nasief H, Mirdad A, Malibary H, Hakamy S, Hassan A, Alaifan M, Bamaga A, Shahzad SA. Comprehensive Analysis of Genes Associated With Sudden Infant Death Syndrome. Front Pediatr 2021; 9:742225. [PMID: 34722422 PMCID: PMC8555024 DOI: 10.3389/fped.2021.742225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/13/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Sudden infant death syndrome (SIDS) is a tragic incident which remains a mystery even after post-mortem investigation and thorough researches. Methods: This comprehensive review is based on the genes reported in the molecular autopsy studies conducted on SIDS so far. A total of 20 original studies and 7 case reports were identified and included in this analysis. The genes identified in children or adults were not included. Most of the genes reported in these studies belonged to cardiac channel and cardiomyopathy. Cardiac channel genes in SIDS were scrutinized for further analysis. Results: After screening and removing the duplicates, 42 unique genes were extracted. When the location of these genes was assessed, it was observed that most of these belonged to Chromosomes 11, 1 and 3 in sequential manner. The pathway analysis shows that these genes are involved in the regulation of heart rate, action potential, cardiac muscle cell contraction and heart contraction. The protein-protein interaction network was also very big and highly interactive. SCN5A, CAV3, ALG10B, AKAP9 and many more were mainly found in these cases and were regulated by many transcription factors such as MYOG C2C1 and CBX3 HCT11. Micro RNA, "hsa-miR-133a-3p" was found to be prevalent in the targeted genes. Conclusions: Molecular and computational approaches are a step forward toward exploration of these sad demises. It is so far a new arena but seems promising to dig out the genetic cause of SIDS in the years to come.
Collapse
Affiliation(s)
- Riffat Mehboob
- Research Unit, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan.,Lahore Medical Research Center, LLP, Lahore, Pakistan
| | - Maher Kurdi
- Department of Pathology, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mursleen Ahmad
- Department of Medicine, Sahiwal Medical College, Sahiwal, Pakistan
| | - Syed Amir Gilani
- Research Unit, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Sidra Khalid
- Lahore Medical Research Center, LLP, Lahore, Pakistan
| | - Hisham Nasief
- Department of Obstetric and Gynecology, Faculty of Medicine, King Abdulaziz University and Hospital, Jeddah, Saudi Arabia
| | - Abeer Mirdad
- Pediatric Department, East Jeddah Hospital, Jeddah, Saudi Arabia
| | - Husam Malibary
- Department of Internal Medicine, Faculty of Medicine, King Abdul Aziz University, Jeddah, Saudi Arabia
| | - Sahar Hakamy
- Center of Excellence in Genomic Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Amber Hassan
- Research Unit, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Meshari Alaifan
- Department of Paediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Bamaga
- Paediatric Department, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia.,Neurology and Pediatric Department, Faculty of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Syed Adnan Shahzad
- Faculty of Medicine and University Hospital of Cologne, Institute of Virology, University of Cologne, Cologne, Germany
| |
Collapse
|
5
|
Zluhan-Martínez E, Pérez-Koldenkova V, Ponce-Castañeda MV, Sánchez MDLP, García-Ponce B, Miguel-Hernández S, Álvarez-Buylla ER, Garay-Arroyo A. Beyond What Your Retina Can See: Similarities of Retinoblastoma Function between Plants and Animals, from Developmental Processes to Epigenetic Regulation. Int J Mol Sci 2020; 21:E4925. [PMID: 32664691 PMCID: PMC7404004 DOI: 10.3390/ijms21144925] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
The Retinoblastoma protein (pRb) is a key cell cycle regulator conserved in a wide variety of organisms. Experimental analysis of pRb's functions in animals and plants has revealed that this protein participates in cell proliferation and differentiation processes. In addition, pRb in animals and its orthologs in plants (RBR), are part of highly conserved protein complexes which suggest the possibility that analogies exist not only between functions carried out by pRb orthologs themselves, but also in the structure and roles of the protein networks where these proteins are involved. Here, we present examples of pRb/RBR participation in cell cycle control, cell differentiation, and in the regulation of epigenetic changes and chromatin remodeling machinery, highlighting the similarities that exist between the composition of such networks in plants and animals.
Collapse
Affiliation(s)
- Estephania Zluhan-Martínez
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de Mexico, 3er Circuito Ext. Junto a J. Botánico, Ciudad Universitaria, UNAM 04510, Mexico; (E.Z.-M.); (M.d.l.P.S.); (B.G.-P.)
- Posgrado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, Av. Universidad 3000, Coyoacán 04510, Mexico
| | - Vadim Pérez-Koldenkova
- Laboratorio Nacional de Microscopía Avanzada, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Av. Cuauhtémoc, 330. Col. Doctores, Alc. Cuauhtémoc 06720, Mexico;
| | - Martha Verónica Ponce-Castañeda
- Unidad de Investigación Médica en Enfermedades Infecciosas, Centro Médico Nacional SXXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - María de la Paz Sánchez
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de Mexico, 3er Circuito Ext. Junto a J. Botánico, Ciudad Universitaria, UNAM 04510, Mexico; (E.Z.-M.); (M.d.l.P.S.); (B.G.-P.)
| | - Berenice García-Ponce
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de Mexico, 3er Circuito Ext. Junto a J. Botánico, Ciudad Universitaria, UNAM 04510, Mexico; (E.Z.-M.); (M.d.l.P.S.); (B.G.-P.)
| | - Sergio Miguel-Hernández
- Laboratorio de Citopatología Ambiental, Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Campus Zacatenco, Calle Wilfrido Massieu Esquina Cda, Manuel Stampa 07738, Mexico;
| | - Elena R. Álvarez-Buylla
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de Mexico, 3er Circuito Ext. Junto a J. Botánico, Ciudad Universitaria, UNAM 04510, Mexico; (E.Z.-M.); (M.d.l.P.S.); (B.G.-P.)
| | - Adriana Garay-Arroyo
- Laboratorio de Genética Molecular, Epigenética, Desarrollo y Evolución de Plantas, Instituto de Ecología, Universidad Nacional Autónoma de Mexico, 3er Circuito Ext. Junto a J. Botánico, Ciudad Universitaria, UNAM 04510, Mexico; (E.Z.-M.); (M.d.l.P.S.); (B.G.-P.)
| |
Collapse
|
6
|
MyoD1 suppresses cell migration and invasion by inhibiting FUT4 transcription in human gastric cancer cells. Cancer Gene Ther 2019; 27:773-784. [PMID: 31831855 PMCID: PMC7661344 DOI: 10.1038/s41417-019-0153-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023]
Abstract
Myogenic differentiation 1 (MyoD1) is a transcription factor that promotes expression of muscle-specific genes. MyoD1 is expressed at significantly lower levels in gastric cancer (GC) tissues and cells, and it induces apoptosis in GC cells. However, functions for MyoD1 in GC cell migration and gene expression have not been documented. We show that knockdown of MyoD1 promoted migration and invasion of GC cells, whereas MyoD1 overexpression suppressed migration and invasion. We performed chromatin immunoprecipitation (ChIP)-sequencing to identify MyoD1 target genes in MKN-45 cells. The 2-kb upstream regions (Up2k) of the transcription start sites of 57 genes were probably bound by MyoD1. Six of these genes function in signaling pathways such as synthesis of glycosphingolipid biosynthesis—lacto and neolacto series. MyoD1 inhibited transcription of fucosyltransferase IV (FUT4) by binding directly to the FUT4 F3; this finding was validated by ChIP-quantitative PCR and a luciferase reporter assay. Ulex europaeus agglutinin I, which binds Fucα1-2Galβ1-4GlcNAc, and Lewis antigens showed decreased binding to the plasma membrane of cells that overexpressed MyoD1. Knockdown of FUT4 mimicked MyoD1 overexpression by suppressing GC cell migration and invasion; this result implied that MyoD1 suppressed cell migration and invasion via inhibiting the FUT4/matrix metallopeptidase signaling pathway. In summary, this study demonstrated that MyoD1 suppresses migration and invasion of GC cells by directly binding to the F3 region in the FUT4 Up2k and inhibiting FUT4/type II Lewis antigen expression.
Collapse
|
7
|
Repression of ESR1 transcription by MYOD potentiates letrozole-resistance in ERα-positive breast cancer cells. Biochem Biophys Res Commun 2017; 492:425-433. [DOI: 10.1016/j.bbrc.2017.08.082] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/21/2017] [Indexed: 11/15/2022]
|
8
|
Liu X, Yang X, Zhu R, Dai M, Zhu M, Shen Y, Fang H, Sang A, Chen H. Involvement of Fra-1 in Retinal Ganglion Cell Apoptosis in Rat Light-Induced Retina Damage Model. Cell Mol Neurobiol 2017; 37:83-92. [PMID: 27002720 DOI: 10.1007/s10571-016-0346-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 02/08/2016] [Indexed: 02/03/2023]
Abstract
Cell cycle re-entry, in which Fra-1 (transcription factor FOS-related antigen 1) plays an important role, is a key process in neuronal apoptosis. However, the expression and function of Fra-1 in retinal ganglion cell (RGC) apoptosis are unknown. To investigate whether Fra-1 was involved in RGC apoptosis, we performed a light-induced retinal damage model in adult rats. Western blot revealed that up-regulation of Fra-1 expression appeared in retina after light exposure (LE). Immunostaining indicated that increased Fra-1 was mainly expressed in RGCs in retinal ganglion cell layer (GCL) after LE. Co-localization of Fra-1 with active caspase-3 or TUNEL-positive cells in GCL after LE was also detected. In addition, Fra-1 expression increased in parallel with cyclin D1 and phosphorylated mitogen-activated protein kinase p38 (p-p38) expression in retina after LE. Furthermore, Fra-1, cyclin D1, and active caspase-3 protein expression decreased by intravitreal injection of SB203580, a highly selective inhibitor of p38 MAP kinase (p38 MAPK). All these results suggested that Fra-1 may be associated with RGC apoptosis after LE regulated by p38 MAPK through cell cycle re-entry mechanism.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Xiaowei Yang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Rongrong Zhu
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Ming Dai
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Manhui Zhu
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Yuntian Shen
- Department of Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, Jiangsu, China
| | - Hongda Fang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Aimin Sang
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China.
| | - Hui Chen
- Department of Ophthalmology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
9
|
Czerwinska AM, Nowacka J, Aszer M, Gawrzak S, Archacka K, Fogtman A, Iwanicka-Nowicka R, Jańczyk-Ilach K, Koblowska M, Ciemerych MA, Grabowska I. Cell cycle regulation of embryonic stem cells and mouse embryonic fibroblasts lacking functional Pax7. Cell Cycle 2016; 15:2931-2942. [PMID: 27610933 PMCID: PMC5105925 DOI: 10.1080/15384101.2016.1231260] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The transcription factor Pax7 plays a key role during embryonic myogenesis and in adult organisms in that it sustains the proper function of satellite cells, which serve as adult skeletal muscle stem cells. Recently we have shown that lack of Pax7 does not prevent the myogenic differentiation of pluripotent stem cells. In the current work we show that the absence of functional Pax7 in differentiating embryonic stem cells modulates cell cycle facilitating their proliferation. Surprisingly, deregulation of Pax7 function also positively impacts at the proliferation of mouse embryonic fibroblasts. Such phenotypes seem to be executed by modulating the expression of positive cell cycle regulators, such as cyclin E.
Collapse
Affiliation(s)
- Areta M Czerwinska
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Joanna Nowacka
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Magdalena Aszer
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Sylwia Gawrzak
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Karolina Archacka
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Anna Fogtman
- b Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Warsaw , Poland
| | - Roksana Iwanicka-Nowicka
- b Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Warsaw , Poland.,c Department of Systems Biology , Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Katarzyna Jańczyk-Ilach
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Marta Koblowska
- b Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Warsaw , Poland.,c Department of Systems Biology , Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Maria A Ciemerych
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Iwona Grabowska
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| |
Collapse
|
10
|
Huang Y, Chen B, Ye M, Liang P, Zhangfang Y, Huang J, Liu M, Songyang Z, Ma W. Ccndbp1 is a new positive regulator of skeletal myogenesis. J Cell Sci 2016; 129:2767-77. [PMID: 27235421 DOI: 10.1242/jcs.184234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 05/02/2016] [Indexed: 12/26/2022] Open
Abstract
Skeletal myogenesis is a multistep process in which basic helix-loop-helix (bHLH) transcription factors, such as MyoD (also known as MyoD1), bind to E-boxes and activate downstream genes. Ccndbp1 is a HLH protein that lacks a DNA-binding region, and its function in skeletal myogenesis is currently unknown. We generated Ccndbp1-null mice by using CRISPR-Cas9. Notably, in Ccndbp1-null mice, the cross sectional area of the skeletal tibialis anterior muscle was smaller, and muscle regeneration ability and grip strength were impaired, compared with those of wild type. This phenotype resembled that of myofiber hypotrophy in some human myopathies or amyoplasia. Ccndbp1 expression was upregulated during C2C12 myogenesis. Ccndbp1 overexpression promoted myogenesis, whereas knockdown of Ccndbp1 inhibited myogenic differentiation. Co-transfection of Ccndbp1 with MyoD and/or E47 (encoded by TCF3) significantly enhanced E-box-dependent transcription. Furthermore, Ccndbp1 physically associated with MyoD but not E47. These data suggest that Ccndbp1 regulates muscle differentiation by interacting with MyoD and enhancing its binding to target genes. Our study newly identifies Ccndbp1 as a positive modulator of skeletal myogenic differentiation in vivo and in vitro, providing new insights in order to decipher the complex network involved in skeletal myogenic development and related diseases.
Collapse
Affiliation(s)
- Yan Huang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China Collaborative Innovation Center for Cancer Medicine, Guangzhou Key Laboratory of Healthy Aging Research, Sun Yat-sen University, Guangzhou 510006, China
| | - Bohong Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China
| | - Miaoman Ye
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China
| | - Puping Liang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China
| | - Yingnan Zhangfang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China
| | - Junjiu Huang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Zhou Songyang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China Collaborative Innovation Center for Cancer Medicine, Guangzhou Key Laboratory of Healthy Aging Research, Sun Yat-sen University, Guangzhou 510006, China
| | - Wenbin Ma
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 51006, China Collaborative Innovation Center for Cancer Medicine, Guangzhou Key Laboratory of Healthy Aging Research, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
11
|
Cai C, Qin X, Wu Z, Shen Q, Yang W, Zhang S, Duan J, Liang F, Liu C. Inhibitory effect of MyoD on the proliferation of breast cancer cells. Oncol Lett 2016; 11:3589-3596. [PMID: 27284360 PMCID: PMC4887810 DOI: 10.3892/ol.2016.4448] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/24/2016] [Indexed: 12/31/2022] Open
Abstract
Skeletal muscle is rich in lymphatic vessels, with an abundant blood supply, and it is an infrequent site of cancer metastasis. Previous studies have demonstrated that enhanced secretion of MyoD may occur when skeletal muscle is injured or becomes cancerous. It was hypothesized that MyoD may act as an endogenous cytokine to inhibit the proliferation of cancer cells. To verify the possible effect of this protein on tumor cell proliferation, C2C12 mouse skeletal muscle cells and 4T1 mouse breast cancer cells were co-cultured using embedded Transwell plates. Following co-culture, cell cycle analysis revealed that C2C12 muscle cells were able to inhibit the proliferation of the breast cancer cells. Subsequently, MyoD was silenced in C2C12 cells to assess its effect on 4T1 cell proliferation. Following co-culture with MyoD-silenced cells, a 5-ethynyl-20-deoxyuridine assay indicated that MyoD silencing prevented the reduction in proliferation of 4T1 cells induced by untransfected C2C12 cells. In summary, the results indicated that MyoD inhibits the proliferation of breast cancer cells and may be a tumor suppressor factor.
Collapse
Affiliation(s)
- Changjing Cai
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Xiaoqun Qin
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Ziyi Wu
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Qixia Shen
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Wenqian Yang
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Shujun Zhang
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Jinling Duan
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Fenglan Liang
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| | - Chi Liu
- Physiology Department, Xiangya Medical School, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
12
|
Porrello A, Piergentili RB. Contextualizing the Genes Altered in Bladder Neoplasms in Pediatric andTeen Patients Allows Identifying Two Main Classes of Biological ProcessesInvolved and New Potential Therapeutic Targets. Curr Genomics 2016; 17:33-61. [PMID: 27013923 PMCID: PMC4780474 DOI: 10.2174/1389202916666151014222603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/29/2015] [Accepted: 07/08/2015] [Indexed: 12/19/2022] Open
Abstract
Research on bladder neoplasms in pediatric and teen patients (BNPTP) has described 21 genes, which are variously involved in this disease and are mostly responsible for deregulated cell proliferation. However, due to the limited number of publications on this subject, it is still unclear what type of relationships there are among these genes and which are the chances that, while having different molecular functions, they i) act as downstream effector genes of well-known pro- or anti- proliferative stimuli and/or interplay with biochemical pathways having oncological relevance or ii) are specific and, possibly, early biomarkers of these pathologies. A Gene Ontology (GO)-based analysis showed that these 21 genes are involved in biological processes, which can be split into two main classes: cell regulation-based and differentiation/development-based. In order to understand the involvement/overlapping with main cancer-related pathways, we performed a meta-analysis dependent on the 189 oncogenic signatures of the Molecular Signatures Database (OSMSD) curated by the Broad Institute. We generated a binary matrix with 53 gene signatures having at least one hit; this analysis i) suggests that some genes of the original list show inconsistencies and might need to be experimentally re- assessed or evaluated as biomarkers (in particular, ACTA2) and ii) allows hypothesizing that important (proto)oncogenes (E2F3, ERBB2/HER2, CCND1, WNT1, and YAP1) and (putative) tumor suppressors (BRCA1, RBBP8/CTIP, and RB1-RBL2/p130) may participate in the onset of this disease or worsen the observed phenotype, thus expanding the list of possible molecular targets for the treatment of BNPTP.
Collapse
Affiliation(s)
- A. Porrello
- Comprehensive Cancer Center (LCCC), University of North Carolina (UNC)-Chapel Hill, Chapel Hill, 27599 NC, USA
| | - R. b Piergentili
- Institute of Molecular Biology and Pathology at CNR (CNR-IBPM); Department of Biology and Biotechnologies, Sapienza – Università di Roma, Italy
| |
Collapse
|
13
|
Santos-Zas I, Gurriarán-Rodríguez U, Cid-Díaz T, Figueroa G, González-Sánchez J, Bouzo-Lorenzo M, Mosteiro CS, Señarís J, Casanueva FF, Casabiell X, Gallego R, Pazos Y, Mouly V, Camiña JP. β-Arrestin scaffolds and signaling elements essential for the obestatin/GPR39 system that determine the myogenic program in human myoblast cells. Cell Mol Life Sci 2016; 73:617-35. [PMID: 26211463 PMCID: PMC11108386 DOI: 10.1007/s00018-015-1994-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/08/2015] [Accepted: 07/16/2015] [Indexed: 12/27/2022]
Abstract
Obestatin/GPR39 signaling stimulates skeletal muscle repair by inducing the expansion of satellite stem cells as well as myofiber hypertrophy. Here, we describe that the obestatin/GPR39 system acts as autocrine/paracrine factor on human myogenesis. Obestatin regulated multiple steps of myogenesis: myoblast proliferation, cell cycle exit, differentiation and recruitment to fuse and form multinucleated hypertrophic myotubes. Obestatin-induced mitogenic action was mediated by ERK1/2 and JunD activity, being orchestrated by a G-dependent mechanism. At a later stage of myogenesis, scaffolding proteins β-arrestin 1 and 2 were essential for the activation of cell cycle exit and differentiation through the transactivation of the epidermal growth factor receptor (EGFR). Upon obestatin stimulus, β-arrestins are recruited to the membrane, where they functionally interact with GPR39 leading to Src activation and signalplex formation to EGFR transactivation by matrix metalloproteinases. This signalplex regulated the mitotic arrest by p21 and p57 expression and the mid- to late stages of differentiation through JNK/c-Jun, CAMKII, Akt and p38 pathways. This finding not only provides the first functional activity for β-arrestins in myogenesis but also identify potential targets for therapeutic approaches by triggering specific signaling arms of the GPR39 signaling involved in myogenesis.
Collapse
Affiliation(s)
- Icía Santos-Zas
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Uxía Gurriarán-Rodríguez
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- Sprott Centre for Stem Cell Research, Ottawa Health Research Institute, Ottawa, Canada
| | - Tania Cid-Díaz
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Gabriela Figueroa
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
| | - Jessica González-Sánchez
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Mónica Bouzo-Lorenzo
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Carlos S Mosteiro
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - José Señarís
- Servicio de Cirugía Ortopédica y Traumatología, CHUS, SERGAS, Santiago de Compostela, Spain
| | - Felipe F Casanueva
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
- Departamento de Medicina, USC, Santiago de Compostela, Spain
| | - Xesús Casabiell
- Departamento de Fisiología, USC, Santiago de Compostela, Spain
| | - Rosalía Gallego
- Departamento de Ciencias Morfológicas, USC, Santiago de Compostela, Spain
| | - Yolanda Pazos
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Vincent Mouly
- Institut de Myologie, INSERM, and Sorbonne Universités, Université Pierre et Marie Curie, Paris, France
| | - Jesús P Camiña
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain.
| |
Collapse
|
14
|
Bai Y, Li J, Li J, Liu Y, Zhang B. MiR-615 inhibited cell proliferation and cell cycle of human breast cancer cells by suppressing of AKT2 expression. Int J Clin Exp Med 2015; 8:3801-3808. [PMID: 26064277 PMCID: PMC4443111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/25/2015] [Indexed: 06/04/2023]
Abstract
MicroRNAs (miRNAs) have emerged as important regulators that potentially play critical roles in various biological processes. Previous studies have shown that miR-615 regulates proliferation and apoptosis in many types of cancers. The biological function of this microRNA in breast cancer remains largely unexplored. In the present study, we found that miR-615 expression was markedly downregulated in breast cancer tissues and breast cancer cells. The enforced expression of miR-615 was able to inhibite the proliferation and anchorage-independent growth of breast cancer cells, while miR-615-in showed the opposite effect. Bioinformatics analysis further revealed AKT2, a putative tumor promoter as a potential target of miR-615. Ectopic expression of miR-615 led to downregulation of AKT2 protein, which resulted in the upregulation of p27 and p21 and the downregulation of cyclin D1. In sum, these results suggest that miR-615 represents a potential anti-onco-miR and participates in breast cancer carcinogenesis by suppressing AKT2 expression.
Collapse
Affiliation(s)
- Yang Bai
- Department of Surgical Oncology, Cangzhou Centerl HospitalCangzhou, Hebei Province, P. R. China
| | - Jingyan Li
- Department of Radiology, Hebei province Cangzhou Hospital of integrated traditional and Western Medicine (Cangzhou NO. 2 Hospital)Cangzhou, Hebei Province, P. R. China
| | - Jie Li
- Department of Surgical Oncology, Cangzhou Centerl HospitalCangzhou, Hebei Province, P. R. China
| | - Yonghong Liu
- Department of Surgical Oncology, Cangzhou Centerl HospitalCangzhou, Hebei Province, P. R. China
| | - Bo Zhang
- Department of Surgical Oncology, Cangzhou Centerl HospitalCangzhou, Hebei Province, P. R. China
| |
Collapse
|
15
|
MiR-492 contributes to cell proliferation and cell cycle of human breast cancer cells by suppressing SOX7 expression. Tumour Biol 2014; 36:1913-21. [PMID: 25407488 DOI: 10.1007/s13277-014-2794-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 10/30/2014] [Indexed: 10/24/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as important regulators that potentially play critical roles in cancer cell biological processes. Previous studies have shown that miR-492 plays an important role in cell tumorigenesis in multiple kinds of human cancer cells. However, the underlying mechanisms of this microRNA in breast cancer remain largely unknown. In the present study, we investigated miR-492's role in cell proliferation of breast cancer. MiR-492 expression was markedly upregulated in breast cancer tissues and breast cancer cells. Overexpression of miR-492 promoted the proliferation and anchorage-independent growth of breast cancer cells. Bioinformatics analysis further revealed sex-determining region Y-box 7 (SOX7), a putative tumor suppressor, as a potential target of miR-492. Data from luciferase reporter assays showed that miR-492 directly binds to the 3'-untranslated region (3'-UTR) of SOX7 messenger RNA (mRNA) and repressed expression at both transcriptional and translational levels. Ectopic expression of miR-492 led to downregulation of SOX7 protein, which resulted in the upregulation of cyclin D1 and c-Myc. In functional assays, SOX7 silenced in miR-492-in-transfected ZR-75-30 cells has positive effect to promote cell proliferation, suggesting that direct SOX7 downregulation is required for miR-492-induced cell proliferation and cell cycle of breast cancer. In sum, these results suggest that miR-492 represents a potential onco-miR and participates in breast cancer carcinogenesis by suppressing SOX7 expression.
Collapse
|