1
|
Deng Z, Ran Q, Chang W, Li C, Li B, Huang S, Huang J, Zhang K, Li Y, Liu X, Liang Y, Guo Z, Huang S. Cdon is essential for organ left-right patterning by regulating dorsal forerunner cells clustering and Kupffer's vesicle morphogenesis. Front Cell Dev Biol 2024; 12:1429782. [PMID: 39239564 PMCID: PMC11374761 DOI: 10.3389/fcell.2024.1429782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024] Open
Abstract
Cdon and boc are members of the cell adhesion molecule subfamily III Ig/fibronectin. Although they have been reported to be involved in muscle and neural development at late developmental stage, their early roles in embryonic development remain unknown. Here, we discovered that in zebrafish, cdon, but not boc, is expressed in dorsal forerunner cells (DFCs) and the epithelium of Kupffer's vesicle (KV), suggesting a potential role for cdon in organ left-right (LR) patterning. Further data showed that liver and heart LR patterning were disrupted in cdon morphants and cdon mutants. Mechanistically, we found that loss of cdon function led to defect in DFCs clustering, reduced KV lumen, and defective cilia, resulting in randomized Nodal/spaw signaling and subsequent organ LR patterning defects. Additionally, predominant distribution of a cdon morpholino (MO) in DFCs caused defects in DFC clustering, KV morphogenesis, cilia number/length, Nodal/spaw signaling, and organ LR asymmetry, similar to those observed in cdon morphants and cdon -/- embryos, indicating a cell-autonomous role for cdon in regulating KV formation during LR patterning. In conclusion, our data demonstrate that during gastrulation and early somitogenesis, cdon is essential for proper DFC clustering, KV formation, and normal cilia, thereby playing a critical role in establishing organ LR asymmetry.
Collapse
Affiliation(s)
- Zhilin Deng
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
- Department of Ultrasound, Luzhou People's Hospital, Luzhou, China
| | - Qin Ran
- Department of Cardiology, Chengdu Seventh People's Hospital, Chengdu, Sichuan, China
| | - Wenqi Chang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Chengni Li
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Botong Li
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Shuying Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Jingtong Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Ke Zhang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Yuanyuan Li
- Department of Neurology, The Second Affiliated Hospital of Chengdu Medical College, (China National Nuclear Corporation 416 Hospital), Chengdu, China
| | - Xingdong Liu
- Department of Neurology, The Second Affiliated Hospital of Chengdu Medical College, (China National Nuclear Corporation 416 Hospital), Chengdu, China
| | - Yundan Liang
- Department of Pathology and Pathophysiology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Zhenhua Guo
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Sizhou Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
- Department of Neurology, The Second Affiliated Hospital of Chengdu Medical College, (China National Nuclear Corporation 416 Hospital), Chengdu, China
| |
Collapse
|
2
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and machine learning in the prediction and explanation of low pectoralis muscle area. Sci Rep 2024; 14:17981. [PMID: 39097658 PMCID: PMC11297919 DOI: 10.1038/s41598-024-68447-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified eight biomarkers associated with low pectoralis muscle area (PMA). We built three random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified two distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
Affiliation(s)
- Nicholas A Enzer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Joe Chiles
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Stefanie Mason
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Toru Shirahata
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Respiratory Medicine, Saitama Medical University, Saitama, Japan
| | - Victor Castro
- Boston University School of Medicine, Boston, MA, USA
| | - Elizabeth Regan
- COPDGene Study Consortium, Denver, CO, USA
- Division of Rheumatology, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Bina Choi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy F Yuan
- Department of Biomedical Informatics, University of California at San Diego, San Diego, CA, USA
| | - Alejandro A Diaz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Merry-Lynn McDonald
- Division of Pulmonary, Allergy and Critical Care Medicine, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
- COPDGene Study Consortium, Denver, CO, USA
| | - Raúl San José Estépar
- Applied Chest Imaging Laboratory, Brigham and Women's Hospital, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Samuel Y Ash
- Department of Critical Care Medicine, South Shore Hospital, 55 Fogg Road, South Weymouth, MA, 02190, USA.
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
3
|
Mostafa I, Hibberd MC, Hartman SJ, Hafizur Rahman MH, Mahfuz M, Hasan SMT, Ashorn P, Barratt MJ, Ahmed T, Gordon JI. A microbiota-directed complementary food intervention in 12-18-month-old Bangladeshi children improves linear growth. EBioMedicine 2024; 104:105166. [PMID: 38833839 PMCID: PMC11179573 DOI: 10.1016/j.ebiom.2024.105166] [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: 12/05/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Globally, stunting affects ∼150 million children under five, while wasting affects nearly 50 million. Current interventions have had limited effectiveness in ameliorating long-term sequelae of undernutrition including stunting, cognitive deficits and immune dysfunction. Disrupted development of the gut microbiota has been linked to the pathogenesis of undernutrition, providing potentially new treatment approaches. METHODS 124 Bangladeshi children with moderate acute malnutrition (MAM) enrolled (at 12-18 months) in a previously reported 3-month RCT of a microbiota-directed complementary food (MDCF-2) were followed for two years. Weight and length were monitored by anthropometry, the abundances of bacterial strains were assessed by quantifying metagenome-assembled genomes (MAGs) in serially collected fecal samples and levels of growth-associated proteins were measured in plasma. FINDINGS Children who had received MDCF-2 were significantly less stunted during follow-up than those who received a standard ready-to-use supplementary food (RUSF) [linear mixed-effects model, βtreatment group x study week (95% CI) = 0.002 (0.001, 0.003); P = 0.004]. They also had elevated fecal abundances of Agathobacter faecis, Blautia massiliensis, Lachnospira and Dialister, plus increased levels of a group of 37 plasma proteins (linear model; FDR-adjusted P < 0.1), including IGF-1, neurotrophin receptor NTRK2 and multiple proteins linked to musculoskeletal and CNS development, that persisted for 6-months post-intervention. INTERPRETATION MDCF-2 treatment of Bangladeshi children with MAM, which produced significant improvements in wasting during intervention, also reduced stunting during follow-up. These results suggest that the effectiveness of supplementary foods for undernutrition may be improved by including ingredients that sponsor healthy microbiota-host co-development. FUNDING This work was supported by the BMGF (Grants OPP1134649/INV-000247).
Collapse
Affiliation(s)
- Ishita Mostafa
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh; Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Matthew C Hibberd
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA; The Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Steven J Hartman
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA; The Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Md Hasan Hafizur Rahman
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Mustafa Mahfuz
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh; Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - S M Tafsir Hasan
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Per Ashorn
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Michael J Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA; The Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, 1212, Bangladesh
| | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA; The Newman Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
4
|
Enzer NA, Chiles J, Mason S, Shirahata T, Castro V, Regan E, Choi B, Yuan NF, Diaz AA, Washko GR, McDonald ML, Estépar RSJ, Ash SY. Proteomics and Machine Learning in the Prediction and Explanation of Low Pectoralis Muscle Area. RESEARCH SQUARE 2024:rs.3.rs-3957125. [PMID: 38496412 PMCID: PMC10942559 DOI: 10.21203/rs.3.rs-3957125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Low muscle mass is associated with numerous adverse outcomes independent of other associated comorbid diseases. We aimed to predict and understand an individual's risk for developing low muscle mass using proteomics and machine learning. We identified 8 biomarkers associated with low pectoralis muscle area (PMA). We built 3 random forest classification models that used either clinical measures, feature selected biomarkers, or both to predict development of low PMA. The area under the receiver operating characteristic curve for each model was: clinical-only = 0.646, biomarker-only = 0.740, and combined = 0.744. We displayed the heterogenetic nature of an individual's risk for developing low PMA and identified 2 distinct subtypes of participants who developed low PMA. While additional validation is required, our methods for identifying and understanding individual and group risk for low muscle mass could be used to enable developments in the personalized prevention of low muscle mass.
Collapse
|
5
|
Schweizer RM, Ivy CM, Natarajan C, Scott GR, Storz JF, Cheviron ZA. Gene regulatory changes underlie developmental plasticity in respiration and aerobic performance in highland deer mice. Mol Ecol 2023; 32:3483-3496. [PMID: 37073620 PMCID: PMC10330314 DOI: 10.1111/mec.16953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 04/20/2023]
Abstract
Phenotypic plasticity can play an important role in the ability of animals to tolerate environmental stress, but the nature and magnitude of plastic responses are often specific to the developmental timing of exposure. Here, we examine changes in gene expression in the diaphragm of highland deer mice (Peromyscus maniculatus) in response to hypoxia exposure at different stages of development. In highland deer mice, developmental plasticity in diaphragm function may mediate changes in several respiratory traits that influence aerobic metabolism and performance under hypoxia. We generated RNAseq data from diaphragm tissue of adult deer mice exposed to (1) life-long hypoxia (before conception to adulthood), (2) post-natal hypoxia (birth to adulthood), (3) adult hypoxia (6-8 weeks only during adulthood) or (4) normoxia. We found five suites of co-regulated genes that are differentially expressed in response to hypoxia, but the patterns of differential expression depend on the developmental timing of exposure. We also identified four transcriptional modules that are associated with important respiratory traits. Many of the genes in these transcriptional modules bear signatures of altitude-related selection, providing an indirect line of evidence that observed changes in gene expression may be adaptive in hypoxic environments. Our results demonstrate the importance of developmental stage in determining the phenotypic response to environmental stressors.
Collapse
Affiliation(s)
- Rena M. Schweizer
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Catherine M. Ivy
- Department of Biology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | | | - Graham R. Scott
- Department of Biology, McMaster University, Hamilton, ON, Canada, L8S 4K1
| | - Jay F. Storz
- School of Biological Sciences, University of Nebraska, Lincoln, NE 68588, USA
| | - Zachary A. Cheviron
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
6
|
Ahn BY, Jeong Y, Kim S, Zhang Y, Kim SW, Leem YE, Kang JS. Cdon suppresses vascular smooth muscle calcification via repression of the Wnt/Runx2 Axis. Exp Mol Med 2023; 55:120-131. [PMID: 36609601 PMCID: PMC9898282 DOI: 10.1038/s12276-022-00909-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/26/2022] [Accepted: 11/08/2022] [Indexed: 01/09/2023] Open
Abstract
Osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) is a risk factor associated with vascular diseases. Wnt signaling is one of the major mechanisms implicated in the osteogenic conversion of VSMCs. Since Cdon has a negative effect on Wnt signaling in distinct cellular processes, we sought to investigate the role of Cdon in vascular calcification. The expression of Cdon was significantly downregulated in VSMCs of the aortas of patients with atherosclerosis and aortic stenosis. Consistently, calcification models, including vitamin D3 (VD3)-injected mice and VSMCs cultured with calcifying media, exhibited reduced Cdon expression. Cdon ablation mice (cKO) exhibited exacerbated aortic stiffness and calcification in response to VD3 compared to the controls. Cdon depletion induced the osteogenic conversion of VSMCs accompanied by cellular senescence. The Cdon-deficient aortas showed a significant alteration in gene expression related to cell proliferation and differentiation together with Wnt signaling regulators. Consistently, Cdon depletion or overexpression in VSMCs elevated or attenuated Wnt-reporter activities, respectively. The deletion mutant of the second immunoglobulin domain (Ig2) in the Cdon ectodomain failed to suppress Wnt signaling and osteogenic conversion of VSMCs. Furthermore, treatment with purified recombinant proteins of the entire ectodomain or Ig2 domain of Cdon displayed suppressive effects on Wnt signaling and VSMC calcification. Our results demonstrate a protective role of Cdon in VSMC calcification by suppressing Wnt signaling. The Ig2 domain of Cdon has the potential as a therapeutic tool to prevent vascular calcification.
Collapse
Affiliation(s)
- Byeong-Yun Ahn
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, School of Medicine, Suwon, South Korea
| | - Yideul Jeong
- Research Institute of Aging Related Disease, AniMusCure, Inc., Suwon, South Korea
| | - Sunghee Kim
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, School of Medicine, Suwon, South Korea
| | - Yan Zhang
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, School of Medicine, Suwon, South Korea
| | - Su Woo Kim
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, School of Medicine, Suwon, South Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, School of Medicine, Suwon, South Korea.
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Single Cell Network Research Center, Sungkyunkwan University, School of Medicine, Suwon, South Korea.
| |
Collapse
|
7
|
Hwang J, Thurmond DC. Exocytosis Proteins: Typical and Atypical Mechanisms of Action in Skeletal Muscle. Front Endocrinol (Lausanne) 2022; 13:915509. [PMID: 35774142 PMCID: PMC9238359 DOI: 10.3389/fendo.2022.915509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Insulin-stimulated glucose uptake in skeletal muscle is of fundamental importance to prevent postprandial hyperglycemia, and long-term deficits in insulin-stimulated glucose uptake underlie insulin resistance and type 2 diabetes. Skeletal muscle is responsible for ~80% of the peripheral glucose uptake from circulation via the insulin-responsive glucose transporter GLUT4. GLUT4 is mainly sequestered in intracellular GLUT4 storage vesicles in the basal state. In response to insulin, the GLUT4 storage vesicles rapidly translocate to the plasma membrane, where they undergo vesicle docking, priming, and fusion via the high-affinity interactions among the soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) exocytosis proteins and their regulators. Numerous studies have elucidated that GLUT4 translocation is defective in insulin resistance and type 2 diabetes. Emerging evidence also links defects in several SNAREs and SNARE regulatory proteins to insulin resistance and type 2 diabetes in rodents and humans. Therefore, we highlight the latest research on the role of SNAREs and their regulatory proteins in insulin-stimulated GLUT4 translocation in skeletal muscle. Subsequently, we discuss the novel emerging role of SNARE proteins as interaction partners in pathways not typically thought to involve SNAREs and how these atypical functions reveal novel therapeutic targets for combating peripheral insulin resistance and diabetes.
Collapse
Affiliation(s)
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute at City of Hope, Duarte, CA, United States
| |
Collapse
|
8
|
Uluca B, Lektemur Esen C, Saritas Erdogan S, Kumbasar A. NFI transcriptionally represses CDON and is required for SH-SY5Y cell survival. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194798. [PMID: 35151899 DOI: 10.1016/j.bbagrm.2022.194798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
Nuclear Factor One (NFI) family of transcription factors regulate proliferation and multiple aspects of differentiation, playing analogous roles in embryonic development and various types of cancer. While all NFI family members are expressed in the developing brain and are involved in progression of brain cancers, their role in neuroblastoma has not been studied. Here we show that NFIB is required for the survival and proliferation of SH-SY5Y neuroblastoma cells, assessed by viability and colony formation assays. Cdon, an Ig superfamily member, is a SHH dependence receptor that acts as a tumor suppressor in neuroblastoma. In the absence of NFI, Cdon is upregulated in the developing mouse brain, however the mechanisms by which its transcription is regulated remains unknown. We report CDON as a downstream target of NFIs in SH-SY5Y cells. There are three putative NFI binding sites within the one kb CDON promoter, two of which are occupied by NFIs in SH-SY5Y cells and human neural stem cells. In dual-luciferase assays, Nfib directly represses CDON proximal promoter activity. Moreover, silencing NFIB leads to upregulation of CDON in SH-SY5Y cells, however, decreased cell proliferation in NFIB silenced cells could not be rescued by concomitantly silencing CDON, suggesting other molecular players are involved. For instance, p21, an NFI target in glioblastoma and breast cancer cells, is also upregulated upon NFIB knock-down. We propose that NFIB is indispensable for SH-SY5Y cells which may involve regulation of apoptosis inducer proteins CDON and p21.
Collapse
Affiliation(s)
- Betül Uluca
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; Department of Molecular Biotechnology, Turkish-German University, Beykoz, Istanbul 34820, Turkey
| | - Cemre Lektemur Esen
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey
| | - Sinem Saritas Erdogan
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey
| | - Asli Kumbasar
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey.
| |
Collapse
|
9
|
Taylor L, Wankell M, Saxena P, McFarlane C, Hebbard L. Cell adhesion an important determinant of myogenesis and satellite cell activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119170. [PMID: 34763027 DOI: 10.1016/j.bbamcr.2021.119170] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/18/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022]
Abstract
Skeletal muscles represent a complex and highly organised tissue responsible for all voluntary body movements. Developed through an intricate and tightly controlled process known as myogenesis, muscles form early in development and are maintained throughout life. Due to the constant stresses that muscles are subjected to, skeletal muscles maintain a complex course of regeneration to both replace and repair damaged myofibers and to form new functional myofibers. This process, made possible by a pool of resident muscle stem cells, termed satellite cells, and controlled by an array of transcription factors, is additionally reliant on a diverse range of cell adhesion molecules and the numerous signaling cascades that they initiate. This article will review the literature surrounding adhesion molecules and their roles in skeletal muscle myogenesis and repair.
Collapse
Affiliation(s)
- Lauren Taylor
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia
| | - Miriam Wankell
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia
| | - Pankaj Saxena
- Department of Cardiothoracic Surgery, The Townsville University Hospital, Townsville, Queensland, Australia; College of Medicine, Dentistry, James Cook University, Townsville, Queensland, Australia
| | - Craig McFarlane
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia.
| | - Lionel Hebbard
- Department of Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, Centre for Molecular Therapeutics, Centre for Tropical Bioinformatics and Molecular Biology, Australian Institute of Tropical Medicine and Health, James Cook University, Townsville, Queensland, Australia; Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
10
|
ZSWIM8 is a myogenic protein that partly prevents C2C12 differentiation. Sci Rep 2021; 11:20880. [PMID: 34686700 PMCID: PMC8536758 DOI: 10.1038/s41598-021-00306-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
Cell adhesion molecule-related/downregulated by oncogenes (Cdon) is a cell-surface receptor that mediates cell–cell interactions and positively regulates myogenesis. The cytoplasmic region of Cdon interacts with other proteins to form a Cdon/JLP/Bnip-2/CDC42 complex that activates p38 mitogen-activated protein kinase (MAPK) and induces myogenesis. However, Cdon complex may include other proteins during myogenesis. In this study, we found that Cullin 2-interacting protein zinc finger SWIM type containing 8 (ZSWIM8) ubiquitin ligase is induced during C2C12 differentiation and is included in the Cdon complex. We knocked-down Zswim8 in C2C12 cells to determine the effect of ZSWIM8 on differentiation. However, we detected neither ZSWIM8-dependent ubiquitination nor the degradation of Bnip2, Cdon, or JLP. In contrast, ZSWIM8 knockdown accelerated C2C12 differentiation. These results suggest that ZSWIM8 is a Cdon complex-included myogenic protein that prevents C2C12 differentiation without affecting the stability of Bnip2, Cdon, and JLP.
Collapse
|
11
|
Malacarne C, Galbiati M, Giagnorio E, Cavalcante P, Salerno F, Andreetta F, Cagnoli C, Taiana M, Nizzardo M, Corti S, Pensato V, Venerando A, Gellera C, Fenu S, Pareyson D, Masson R, Maggi L, Dalla Bella E, Lauria G, Mantegazza R, Bernasconi P, Poletti A, Bonanno S, Marcuzzo S. Dysregulation of Muscle-Specific MicroRNAs as Common Pathogenic Feature Associated with Muscle Atrophy in ALS, SMA and SBMA: Evidence from Animal Models and Human Patients. Int J Mol Sci 2021; 22:ijms22115673. [PMID: 34073630 PMCID: PMC8198536 DOI: 10.3390/ijms22115673] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/20/2021] [Accepted: 05/23/2021] [Indexed: 02/07/2023] Open
Abstract
Motor neuron diseases (MNDs) are neurodegenerative disorders characterized by upper and/or lower MN loss. MNDs include amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA), and spinal and bulbar muscular atrophy (SBMA). Despite variability in onset, progression, and genetics, they share a common skeletal muscle involvement, suggesting that it could be a primary site for MND pathogenesis. Due to the key role of muscle-specific microRNAs (myomiRs) in skeletal muscle development, by real-time PCR we investigated the expression of miR-206, miR-133a, miR-133b, and miR-1, and their target genes, in G93A-SOD1 ALS, Δ7SMA, and KI-SBMA mouse muscle during disease progression. Further, we analyzed their expression in serum of SOD1-mutated ALS, SMA, and SBMA patients, to demonstrate myomiR role as noninvasive biomarkers. Our data showed a dysregulation of myomiRs and their targets, in ALS, SMA, and SBMA mice, revealing a common pathogenic feature associated with muscle impairment. A similar myomiR signature was observed in patients’ sera. In particular, an up-regulation of miR-206 was identified in both mouse muscle and serum of human patients. Our overall findings highlight the role of myomiRs as promising biomarkers in ALS, SMA, and SBMA. Further investigations are needed to explore the potential of myomiRs as therapeutic targets for MND treatment.
Collapse
Affiliation(s)
- Claudia Malacarne
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
- PhD Program in Neuroscience, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy;
| | - Eleonora Giagnorio
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
- PhD Program in Neuroscience, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Paola Cavalcante
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
| | - Franco Salerno
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
| | - Francesca Andreetta
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
| | - Cinza Cagnoli
- Molecular Neuroanatomy and Pathogenesis Unit, Neurology VII—Clinical and Experimental Epileptology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy;
| | - Michela Taiana
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; (M.T.); (S.C.)
| | - Monica Nizzardo
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy;
| | - Stefania Corti
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Via Francesco Sforza 35, 20122 Milan, Italy; (M.T.); (S.C.)
- Neurology Unit, IRCCS Foundation Ca’ Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20122 Milan, Italy;
| | - Viviana Pensato
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (V.P.); (A.V.); (C.G.)
| | - Anna Venerando
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (V.P.); (A.V.); (C.G.)
| | - Cinzia Gellera
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (V.P.); (A.V.); (C.G.)
| | - Silvia Fenu
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (S.F.); (D.P.)
| | - Davide Pareyson
- Unit of Rare Neurodegenerative and Neurometabolic Diseases, Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (S.F.); (D.P.)
| | - Riccardo Masson
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy;
| | - Lorenzo Maggi
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
| | - Eleonora Dalla Bella
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (E.D.B.); (G.L.)
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (E.D.B.); (G.L.)
- Department of Biomedical and Clinical Sciences “Luigi Sacco”, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Renato Mantegazza
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
| | - Pia Bernasconi
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Via Balzaretti, 9, 20133 Milano, Italy;
- Correspondence: (A.P.); (S.M.); Tel.: +39-02-5031-8215 (A.P.); Tel.: +39-02-2394-4511 (ext. 4651) (S.M.); Fax: +39-02-70633874 (S.M.)
| | - Silvia Bonanno
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
| | - Stefania Marcuzzo
- Neurology IV–Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (C.M.); (E.G.); (P.C.); (F.S.); (F.A.); (L.M.); (R.M.); (P.B.); (S.B.)
- Correspondence: (A.P.); (S.M.); Tel.: +39-02-5031-8215 (A.P.); Tel.: +39-02-2394-4511 (ext. 4651) (S.M.); Fax: +39-02-70633874 (S.M.)
| |
Collapse
|
12
|
Abundant Synthesis of Netrin-1 in Satellite Cell-Derived Myoblasts Isolated from EDL Rather Than Soleus Muscle Regulates Fast-Type Myotube Formation. Int J Mol Sci 2021; 22:ijms22094499. [PMID: 33925862 PMCID: PMC8123454 DOI: 10.3390/ijms22094499] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/23/2021] [Indexed: 01/05/2023] Open
Abstract
Resident myogenic stem cells (satellite cells) are attracting attention for their novel roles in myofiber type regulation. In the myogenic differentiation phase, satellite cells from soleus muscle (slow fiber-abundant) synthesize and secrete higher levels of semaphorin 3A (Sema3A, a multifunctional modulator) than those derived from extensor digitorum longus (EDL; fast fiber-abundant), suggesting the role of Sema3A in forming slow-twitch myofibers. However, the regulatory mechanisms underlying fast-twitch myotube commitment remain unclear. Herein, we focused on netrin family members (netrin-1, -3, and -4) that compete with Sema3A in neurogenesis and osteogenesis. We examined whether netrins affect fast-twitch myotube generation by evaluating their expression in primary satellite cell cultures. Initially, netrins are upregulated during myogenic differentiation. Next, we compared the expression levels of netrins and their cell membrane receptors between soleus- and EDL-derived satellite cells; only netrin-1 showed higher expression in EDL-derived satellite cells than in soleus-derived satellite cells. We also performed netrin-1 knockdown experiments and additional experiments with recombinant netrin-1 in differentiated satellite cell-derived myoblasts. Netrin-1 knockdown in myoblasts substantially reduced fast-type myosin heavy chain (MyHC) expression; exogenous netrin-1 upregulated fast-type MyHC in satellite cells. Thus, netrin-1 synthesized in EDL-derived satellite cells may promote myofiber type commitment of fast muscles.
Collapse
|
13
|
The cooperation of cis-elements during M-cadherin promoter activation. Biochem J 2021; 478:911-926. [PMID: 33527978 DOI: 10.1042/bcj20200535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 01/21/2021] [Accepted: 02/02/2021] [Indexed: 01/26/2023]
Abstract
M-cadherin is a skeletal muscle-specific transmembrane protein mediating the cell-cell adhesion of myoblasts during myogenesis. It is expressed in the proliferating satellite cells and highly induced by myogenic regulatory factors (MRFs) during terminal myogenic differentiation. Several conserved cis-elements, including 5 E-boxes, 2 GC boxes, and 1 conserved downstream element (CDE) were identified in the M-cadherin proximal promoter. We found that E-box-3 and -4 close to the transcription initiation site (TIS) mediated most of its transactivation by MyoD, the strongest myogenic MRF. Including of any one of the other E-boxes restored the full activation by MyoD, suggesting an essential collaboration between E-boxes. Stronger activation of M-cadherin promoter than that of muscle creatine kinase (MCK) by MyoD was observed regardless of culture conditions and the presence of E47. Furthermore, MyoD/E47 heterodimer and MyoD ∼ E47 fusion protein achieved similar levels of activation in differentiation medium (DM), suggesting high affinity of MyoD/E47 to E-boxes 3/4 under DM. We also found that GC boxes and CDE positively affected MyoD mediated activation. The CDE element was predicted to be the target of the chromatin-modifying factor Meis1/Pbx1 heterodimer. Knockdown of Pbx1 significantly reduced the expression level of M-cadherin, but increased that of N-cadherin. Using ChIP assay, we further found significant reduction in MyoD recruitment to M-cadherin promoter when CDE was deleted. Taken together, these observations suggest that the chromatin-modifying function of Pbx1/Meis1 is critical to M-cadherin promoter activation before MyoD is recruited to E-boxes to trigger transcription.
Collapse
|
14
|
Chen X, Sun Y, Zhang T, Roepstorff P, Yang F. Comprehensive Analysis of the Proteome and PTMomes of C2C12 Myoblasts Reveals that Sialylation Plays a Role in the Differentiation of Skeletal Muscle Cells. J Proteome Res 2020; 20:222-235. [PMID: 33216553 DOI: 10.1021/acs.jproteome.0c00353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The C2C12 myoblast is a model that has been used extensively to study the process of skeletal muscle differentiation. Proteomics has advanced our understanding of skeletal muscle biology and also the differentiation process of skeletal muscle cells. However, there is still no comprehensive analysis of C2C12 myoblast proteomes, which is important for the understanding of key drivers for the differentiation of skeletal muscle cells. Here, we conducted multidimensional proteome profiling to get a comprehensive analysis of proteomes and PTMomes of C2C12 myoblasts with a TiSH strategy. A total of 8313 protein groups were identified, including 7827 protein groups from nonmodified peptides, 3803 phosphoproteins, and 977 formerly sialylated N-linked glycoproteins. Integrated analysis of proteomic and PTMomic data showed that almost all of the kinases and transcription factors in the muscle cell differentiation pathway were phosphorylated. Further analysis indicated that sialylation might play a role in the differentiation of C2C12 myoblasts. Further functional analysis demonstrated that C2C12 myoblasts showed a decreased level of sialylation during skeletal muscle cell differentiation. Inhibition of sialylation with the sialyltransferase inhibitor 3Fax-Neu5Ac resulted in the lower expression of MHC and suppression of myoblast fusion. In all, these results indicate that sialylation has an effect on the differentiation of skeletal muscle cells.
Collapse
Affiliation(s)
- Xiulan Chen
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100149, China
| | - Yaping Sun
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100149, China
| | - Tingting Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100149, China
| | - Peter Roepstorff
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Fuquan Yang
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100149, China
| |
Collapse
|
15
|
Bae JH, Hong M, Jeong HJ, Kim H, Lee SJ, Ryu D, Bae GU, Cho SC, Lee YS, Krauss RS, Kang JS. Satellite cell-specific ablation of Cdon impairs integrin activation, FGF signalling, and muscle regeneration. J Cachexia Sarcopenia Muscle 2020; 11:1089-1103. [PMID: 32103583 PMCID: PMC7432598 DOI: 10.1002/jcsm.12563] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 12/04/2019] [Accepted: 02/09/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Perturbation in cell adhesion and growth factor signalling in satellite cells results in decreased muscle regenerative capacity. Cdon (also called Cdo) is a component of cell adhesion complexes implicated in myogenic differentiation, but its role in muscle regeneration remains to be determined. METHODS We generated inducible satellite cell-specific Cdon ablation in mice by utilizing a conditional Cdon allele and Pax7 CreERT2 . To induce Cdon ablation, mice were intraperitoneally injected with tamoxifen (tmx). Using cardiotoxin-induced muscle injury, the effect of Cdon depletion on satellite cell function was examined by histochemistry, immunostaining, and 5-ethynyl-2'-deoxyuridine (EdU) incorporation assay. Isolated myofibers or myoblasts were utilized to determine stem cell function and senescence. To determine pathways related to Cdon deletion, injured muscles were subjected to RNA sequencing analysis. RESULTS Satellite cell-specific Cdon ablation causes impaired muscle regeneration with fibrosis, likely attributable to decreased proliferation, and senescence, of satellite cells. Cultured Cdon-depleted myofibers exhibited 32 ± 9.6% of EdU-positive satellite cells compared with 58 ± 4.4% satellite cells in control myofibers (P < 0.05). About 32.5 ± 3.7% Cdon-ablated myoblasts were positive for senescence-associated β-galactosidase (SA-β-gal) while only 3.6 ± 0.5% of control satellite cells were positive (P < 0.001). Transcriptome analysis of muscles at post-injury Day 4 revealed alterations in genes related to mitogen-activated protein kinase signalling (P < 8.29 e-5 ) and extracellular matrix (P < 2.65 e-24 ). Consistent with this, Cdon-depleted tibialis anterior muscles had reduced phosphorylated extracellular signal-regulated kinase (p-ERK) protein levels and expression of ERK targets, such as Fos (0.23-fold) and Egr1 (0.31-fold), relative to mock-treated control muscles (P < 0.001). Cdon-depleted myoblasts exhibited impaired ERK activation in response to basic fibroblast growth factor. Cdon ablation resulted in decreased and/or mislocalized integrin β1 activation in satellite cells (weak or mislocalized integrin1 in tmx = 38.7 ± 1.9%, mock = 21.5 ± 6%, P < 0.05), previously linked with reduced fibroblast growth factor (FGF) responsiveness in aged satellite cells. In mechanistic studies, Cdon interacted with and regulated cell surface localization of FGFR1 and FGFR4, likely contributing to FGF responsiveness of satellite cells. Satellite cells from a progeria model, Zmpste24-/- myofibers, showed decreased Cdon levels (Cdon-positive cells in Zmpste24-/- = 63.3 ± 11%, wild type = 90 ± 7.7%, P < 0.05) and integrin β1 activation (weak or mislocalized integrin β1 in Zmpste24-/- = 64 ± 6.9%, wild type = 17.4 ± 5.9%, P < 0.01). CONCLUSIONS Cdon deficiency in satellite cells causes impaired proliferation of satellite cells and muscle regeneration via aberrant integrin and FGFR signalling.
Collapse
Affiliation(s)
- Ju-Hyeon Bae
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Mingi Hong
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hyeon-Ju Jeong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Hyebeen Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Sang-Jin Lee
- Research Institute of Pharmaceutical Science, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Republic of Korea
| | - Gyu-Un Bae
- Research Institute of Pharmaceutical Science, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Sung Chun Cho
- Well Aging Research Center, DGIST, Daegu, Republic of Korea
| | - Young-Sam Lee
- Well Aging Research Center, DGIST, Daegu, Republic of Korea.,Department of New Biology, DGIST, Daegu, Republic of Korea
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.,Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
16
|
Jeong HJ, Lee SJ, Lee HJ, Kim HB, Anh Vuong T, Cho H, Bae GU, Kang JS. Prmt7 promotes myoblast differentiation via methylation of p38MAPK on arginine residue 70. Cell Death Differ 2019; 27:573-586. [PMID: 31243342 PMCID: PMC7206020 DOI: 10.1038/s41418-019-0373-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 05/13/2019] [Accepted: 05/19/2019] [Indexed: 01/12/2023] Open
Abstract
MyoD functions as a master regulator to induce muscle-specific gene expression and myogenic differentiation. Here, we demonstrate a positive role of Protein arginine methyltransferase 7 (Prmt7) in MyoD-mediated myoblast differentiation through p38MAPK activation. Prmt7 depletion in primary or C2C12 myoblasts impairs cell cycle withdrawal and myogenic differentiation. Furthermore, Prmt7 depletion decreases the MyoD-reporter activities and the MyoD-mediated myogenic conversion of fibroblasts. Together with MyoD, Prmt7 is recruited to the Myogenin promoter region and Prmt7 depletion attenuates the recruitment of MyoD and its coactivators. The mechanistic study reveals that Prmt7 methylates p38MAPKα at the arginine residue 70, thereby promoting its activation which in turn enhances MyoD activities. The arginine residue 70 to alanine mutation in p38MAPKα impedes MyoD/E47 heterodimerization and the recruitment of Prmt7, MyoD and Baf60c to the Myogenin promoter resulting in blunted Myogenin expression. In conclusion, Prmt7 promotes MyoD-mediated myoblast differentiation through methylation of p38MAPKα at arginine residue 70.
Collapse
Affiliation(s)
- Hyeon-Ju Jeong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Single Cell Network Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sang-Jin Lee
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Hye-Jin Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Single Cell Network Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hye-Been Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Single Cell Network Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Tuan Anh Vuong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Single Cell Network Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Single Cell Network Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Gyu-Un Bae
- Drug Information Research Institute, College of Pharmacy, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Single Cell Network Research Center, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
17
|
Castiglioni I, Caccia R, Garcia-Manteiga JM, Ferri G, Caretti G, Molineris I, Nishioka K, Gabellini D. The Trithorax protein Ash1L promotes myoblast fusion by activating Cdon expression. Nat Commun 2018; 9:5026. [PMID: 30487570 PMCID: PMC6262021 DOI: 10.1038/s41467-018-07313-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/24/2018] [Indexed: 12/17/2022] Open
Abstract
Myoblast fusion (MF) is required for muscle growth and repair, and its alteration contributes to muscle diseases. The mechanisms governing this process are incompletely understood, and no epigenetic regulator has been previously described. Ash1L is an epigenetic activator belonging to the Trithorax group of proteins and is involved in FSHD muscular dystrophy, autism and cancer. Its physiological role in skeletal muscle is unknown. Here we report that Ash1L expression is positively correlated with MF and reduced in Duchenne muscular dystrophy. In vivo, ex vivo and in vitro experiments support a selective and evolutionary conserved requirement for Ash1L in MF. RNA- and ChIP-sequencing indicate that Ash1L is required to counteract Polycomb repressive activity to allow activation of selected myogenesis genes, in particular the key MF gene Cdon. Our results promote Ash1L as an important epigenetic regulator of MF and suggest that its activity could be targeted to improve cell therapy for muscle diseases. Myoblast fusion in skeletal muscle is a complex process but how this is regulated is unclear. Here, the authors identify Ash1L, a histone methyltransferase, as modulating myoblast fusion via activation of the myogenesis gene Cdon, and observe decreased Ash1L expression in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Ilaria Castiglioni
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Roberta Caccia
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Jose Manuel Garcia-Manteiga
- Center for Translational Genomics and BioInformatics, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Giulia Ferri
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Giuseppina Caretti
- Department of Biosciences, University of Milan, via Celoria 26, Milano, 20133, Italy
| | - Ivan Molineris
- Center for Translational Genomics and BioInformatics, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy
| | - Kenichi Nishioka
- Department of Biomolecular Sciences, Division of Molecular Genetics and Epigenetics, Faculty of Medicine, Saga University, Saga, Japan.,Laboratory for Developmental Genetics, RIKEN IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Davide Gabellini
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, via Olgettina 60, Milano, 20132, Italy.
| |
Collapse
|
18
|
Goel AJ, Rieder MK, Arnold HH, Radice GL, Krauss RS. Niche Cadherins Control the Quiescence-to-Activation Transition in Muscle Stem Cells. Cell Rep 2018; 21:2236-2250. [PMID: 29166613 DOI: 10.1016/j.celrep.2017.10.102] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/01/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022] Open
Abstract
Many adult stem cells display prolonged quiescence, promoted by cues from their niche. Upon tissue damage, a coordinated transition to the activated state is required because non-physiological breaks in quiescence often lead to stem cell depletion and impaired regeneration. Here, we identify cadherin-mediated adhesion and signaling between muscle stem cells (satellite cells [SCs]) and their myofiber niche as a mechanism that orchestrates the quiescence-to-activation transition. Conditional removal of N-cadherin and M-cadherin in mice leads to a break in SC quiescence, with long-term expansion of a regeneration-proficient SC pool. These SCs have an incomplete disruption of the myofiber-SC adhesive junction and maintain niche residence and cell polarity, yet show properties of SCs in a state of transition from quiescence toward full activation. Among these is nuclear localization of β-catenin, which is necessary for this phenotype. Injury-induced perturbation of niche adhesive junctions is therefore a likely first step in the quiescence-to-activation transition.
Collapse
Affiliation(s)
- Aviva J Goel
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marysia-Kolbe Rieder
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hans-Henning Arnold
- Cell and Molecular Biology, Institute of Zoology, Technical University Braunschweig, 38106 Braunschweig, Germany
| | - Glenn L Radice
- Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
19
|
Cerquone Perpetuini A, Re Cecconi AD, Chiappa M, Martinelli GB, Fuoco C, Desiderio G, Castagnoli L, Gargioli C, Piccirillo R, Cesareni G. Group I Paks support muscle regeneration and counteract cancer-associated muscle atrophy. J Cachexia Sarcopenia Muscle 2018; 9:727-746. [PMID: 29781585 PMCID: PMC6104114 DOI: 10.1002/jcsm.12303] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/02/2018] [Accepted: 03/06/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Skeletal muscle is characterized by an efficient regeneration potential that is often impaired during myopathies. Understanding the molecular players involved in muscle homeostasis and regeneration could help to find new therapies against muscle degenerative disorders. Previous studies revealed that the Ser/Thr kinase p21 protein-activated kinase 1 (Pak1) was specifically down-regulated in the atrophying gastrocnemius of Yoshida hepatoma-bearing rats. In this study, we evaluated the role of group I Paks during cancer-related atrophy and muscle regeneration. METHODS We examined Pak1 expression levels in the mouse Tibialis Anterior muscles during cancer cachexia induced by grafting colon adenocarcinoma C26 cells and in vitro by dexamethasone treatment. We investigated whether the overexpression of Pak1 counteracts muscle wasting in C26-bearing mice and in vitro also during interleukin-6 (IL6)-induced or dexamethasone-induced C2C12 atrophy. Moreover, we analysed the involvement of group I Paks on myogenic differentiation in vivo and in vitro using the group I chemical inhibitor IPA-3. RESULTS We found that Pak1 expression levels are reduced during cancer-induced cachexia in the Tibialis Anterior muscles of colon adenocarcinoma C26-bearing mice and in vitro during dexamethasone-induced myotube atrophy. Electroporation of muscles of C26-bearing mice with plasmids directing the synthesis of PAK1 preserves fiber size in cachectic muscles by restraining the expression of atrogin-1 and MuRF1 and possibly by inducing myogenin expression. Consistently, the overexpression of PAK1 reduces the dexamethasone-induced expression of MuRF1 in myotubes and increases the phospho-FOXO3/FOXO3 ratio. Interestingly, the ectopic expression of PAK1 counteracts atrophy in vitro by restraining the IL6-Stat3 signalling pathway measured in luciferase-based assays and by reducing rates of protein degradation in atrophying myotubes exposed to IL6. On the other hand, we observed that the inhibition of group I Paks has no effect on myotube atrophy in vitro and is associated with impaired muscle regeneration in vivo and in vitro. In fact, we found that mice treated with the group I inhibitor IPA-3 display a delayed recovery from cardiotoxin-induced muscle injury. This is consistent with in vitro experiments showing that IPA-3 impairs myogenin expression and myotube formation in vessel-associated myogenic progenitors, C2C12 myoblasts, and satellite cells. Finally, we observed that IPA-3 reduces p38α/β phosphorylation that is required to proceed through various stages of satellite cells differentiation: activation, asymmetric division, and ultimately myotube formation. CONCLUSIONS Our data provide novel evidence that is consistent with group I Paks playing a central role in the regulation of muscle homeostasis, atrophy and myogenesis.
Collapse
Affiliation(s)
| | - Andrea David Re Cecconi
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Michela Chiappa
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Giulia Benedetta Martinelli
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Claudia Fuoco
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Giovanni Desiderio
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Luisa Castagnoli
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Cesare Gargioli
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| | - Rosanna Piccirillo
- Department of NeurosciencesIRCCS‐Mario Negri Institute for Pharmacological ResearchVia Giuseppe La Masa20156MilanItaly
| | - Gianni Cesareni
- Department of BiologyUniversity of Rome Tor VergataVia della ricerca scientifica00133RomeItaly
| |
Collapse
|
20
|
A transcriptomic study of myogenic differentiation under the overexpression of PPARγ by RNA-Seq. Sci Rep 2017; 7:15308. [PMID: 29127356 PMCID: PMC5681552 DOI: 10.1038/s41598-017-14275-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/06/2017] [Indexed: 12/18/2022] Open
Abstract
To study the cellular and molecular function of peroxisome proliferator-activated receptor γ (PPARγ) in skeletal muscle differentiation, we have generated inducible gain-of-function to overexpress PPARγ in C2C12 myoblasts. In order to identify PPARγ targets, RNA sequencing (RNA-seq) was used to evaluate and quantify the transcriptomes and expression patterns during myogenic differentiation under the overexpression of PPARγ. The formation of myotubes and the expression of muscle-specific myogenic genes such as MyoD and MyoG may be inhibited by PPARγ overexpression. Multiple genes and pathways were significantly involved in this process, including 11 genes such as Fndc9 and Slc14a1 with fundamental change of regulation modes, 9 genes of which were validated by the data of qRT-PCR. Our studies demonstrate that PPARγ would play critical roles on myoblasts differentiation, mediating crosstalk among several pathways and transcription factors. Our data is available in the Gene Expression Omnibus (GEO) database with the accession number as GSE99399.
Collapse
|
21
|
Iyer A, Koch AJ, Holaska JM. Expression Profiling of Differentiating Emerin-Null Myogenic Progenitor Identifies Molecular Pathways Implicated in Their Impaired Differentiation. Cells 2017; 6:cells6040038. [PMID: 29065506 PMCID: PMC5755497 DOI: 10.3390/cells6040038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 11/16/2022] Open
Abstract
Mutations in the gene encoding emerin cause Emery-Dreifuss muscular dystrophy (EDMD), a disorder causing progressive skeletal muscle wasting, irregular heart rhythms and contractures of major tendons. RNA sequencing was performed on differentiating wildtype and emerin-null myogenic progenitors to identify molecular pathways implicated in EDMD, 340 genes were uniquely differentially expressed during the transition from day 0 to day 1 in wildtype cells. 1605 genes were uniquely expressed in emerin-null cells; 1706 genes were shared among both wildtype and emerin-null cells. One thousand and forty-seven transcripts showed differential expression during the transition from day 1 to day 2. Four hundred and thirty-one transcripts showed altered expression in both wildtype and emerin-null cells. Two hundred and ninety-five transcripts were differentially expressed only in emerin-null cells and 321 transcripts were differentially expressed only in wildtype cells. DAVID, STRING and Ingenuity Pathway Analysis identified pathways implicated in impaired emerin-null differentiation, including cell signaling, cell cycle checkpoints, integrin signaling, YAP/TAZ signaling, stem cell differentiation, and multiple muscle development and myogenic differentiation pathways. Functional enrichment analysis showed biological functions associated with the growth of muscle tissue and myogenesis of skeletal muscle were inhibited. The large number of differentially expressed transcripts upon differentiation induction suggests emerin functions during transcriptional reprograming of progenitors to committed myoblasts.
Collapse
Affiliation(s)
- Ashvin Iyer
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia, PA 19104, USA.
| | - Adam J Koch
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA.
| | - James M Holaska
- Department of Biomedical Sciences, Rm 534, Cooper Medical School of Rowan University, 401 South Broadway St., Camden, NJ 08028, USA.
- Department of Pharmaceutical Sciences, University of the Sciences, Philadelphia, PA 19104, USA.
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
22
|
Shin JY, Méndez-López I, Hong M, Wang Y, Tanji K, Wu W, Shugol L, Krauss RS, Dauer WT, Worman HJ. Lamina-associated polypeptide 1 is dispensable for embryonic myogenesis but required for postnatal skeletal muscle growth. Hum Mol Genet 2017; 26:65-78. [PMID: 27798115 DOI: 10.1093/hmg/ddw368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 10/21/2016] [Indexed: 12/17/2022] Open
Abstract
Lamina-associated polypeptide 1 (LAP1) is an integral protein of the inner nuclear membrane that has been implicated in striated muscle maintenance. Mutations in its gene have been linked to muscular dystrophy and cardiomyopathy. As germline deletion of the gene encoding LAP1 is perinatal lethal, we explored its potential role in myogenic differentiation and development by generating a conditional knockout mouse in which the protein is depleted from muscle progenitors at embryonic day 8.5 (Myf5-Lap1CKO mice). Although cultured myoblasts lacking LAP1 demonstrated defective terminal differentiation and altered expression of muscle regulatory factors, embryonic myogenesis and formation of skeletal muscle occurred in both mice with a Lap1 germline deletion and Myf5-Lap1CKO mice. However, skeletal muscle fibres were hypotrophic and their nuclei were morphologically abnormal with a wider perinuclear space than normal myonuclei. Myf5-Lap1CKO mouse skeletal muscle contained fewer satellite cells than normal and these cells had evidence of reduced myogenic potential. Abnormalities in signalling pathways required for postnatal hypertrophic growth were also observed in skeletal muscles of these mice. Our results demonstrate that early embryonic depletion of LAP1 does not impair myogenesis but that it is necessary for postnatal skeletal muscle growth.
Collapse
Affiliation(s)
- Ji-Yeon Shin
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Iván Méndez-López
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Mingi Hong
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuexia Wang
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Kurenai Tanji
- Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Wei Wu
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Leana Shugol
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Robert S Krauss
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William T Dauer
- Department of Neurology.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Howard J Worman
- Department of Medicine.,Department of Pathology and Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
23
|
p38 MAPK activation and H3K4 trimethylation is decreased by lactate in vitro and high intensity resistance training in human skeletal muscle. PLoS One 2017; 12:e0176609. [PMID: 28467493 PMCID: PMC5414990 DOI: 10.1371/journal.pone.0176609] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 04/13/2017] [Indexed: 12/04/2022] Open
Abstract
Exercise induces adaptation of skeletal muscle by acutely modulating intracellular signaling, gene expression, protein turnover and myogenic activation of skeletal muscle stem cells (Satellite cells, SCs). Lactate (La)-induced metabolic stimulation alone has been shown to modify SC proliferation and differentiation. Although the mechanistic basis remains elusive, it was demonstrated that La affects signaling via p38 mitogen activated protein kinase (p38 MAPK) which might contribute to trimethylation of histone 3 lysine 4 (H3K4me3) known to regulate satellite cell proliferation and differentiation. We investigated the effects of La on p38 MAPK and H3K4me3 in a model of activated SCs. Differentiating C2C12 myoblasts were treated with La (20 mM) and samples analysed using qRT-PCR, immunofluorescence, and western blotting. We determined a reduction of p38 MAPK phosphorylation, decreased H3K4me3 and reduced expression of Myf5, myogenin, and myosin heavy chain (MHC) leading to decreased differentiation of La-treated C2C12 cells after 5 days of repeated La treatment. We further investigated whether this regulatory pathway would be affected in human skeletal muscle by the application of two different resistance exercise regimes (RE) associated with distinct metabolic demands and blood La accumulation. Muscle biopsies were obtained 15, 30 min, 1, 4, and 24 h post exercise after moderate intensity RE (STD) vs. high intensity RE (HIT). Consistent with in vitro results, reduced p38 phosphorylation and blunted H3K4me3 were also observed upon metabolically demanding HIT RE in human skeletal muscle. Our data provide evidence that La-accumulation acutely affects p38 MAPK signaling, gene expression and thereby cell differentiation and adaptation in vitro, and likely in vivo.
Collapse
|
24
|
Cdon deficiency causes cardiac remodeling through hyperactivation of WNT/β-catenin signaling. Proc Natl Acad Sci U S A 2017; 114:E1345-E1354. [PMID: 28154134 DOI: 10.1073/pnas.1615105114] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
On pathological stress, Wnt signaling is reactivated and induces genes associated with cardiac remodeling and fibrosis. We have previously shown that a cell surface receptor Cdon (cell-adhesion associated, oncogene regulated) suppresses Wnt signaling to promote neuronal differentiation however its role in heart is unknown. Here, we demonstrate a critical role of Cdon in cardiac function and remodeling. Cdon is expressed and predominantly localized at intercalated disk in both mouse and human hearts. Cdon-deficient mice develop cardiac dysfunction including reduced ejection fraction and ECG abnormalities. Cdon-/- hearts exhibit increased fibrosis and up-regulation of genes associated with cardiac remodeling and fibrosis. Electrical remodeling was demonstrated by up-regulation and mislocalization of the gap junction protein, Connexin 43 (Cx43) in Cdon-/- hearts. In agreement with altered Cx43 expression, functional analysis both using Cdon-/- cardiomyocytes and shRNA-mediated knockdown in rat cardiomyocytes shows aberrant gap junction activities. Analysis of the underlying mechanism reveals that Cdon-/- hearts exhibit hyperactive Wnt signaling as evident by β-catenin accumulation and Axin2 up-regulation. On the other hand, the treatment of rat cardiomyocytes with a Wnt activator TWS119 reduces Cdon levels and aberrant Cx43 activities, similarly to Cdon-deficient cardiomyocytes, suggesting a negative feedback between Cdon and Wnt signaling. Finally, inhibition of Wnt/β-catenin signaling by XAV939, IWP2 or dickkopf (DKK)1 prevented Cdon depletion-induced up-regulation of collagen 1a and Cx43. Taken together, these results demonstrate that Cdon deficiency causes hyperactive Wnt signaling leading to aberrant intercellular coupling and cardiac fibrosis. Cdon exhibits great potential as a target for the treatment of cardiac fibrosis and cardiomyopathy.
Collapse
|
25
|
Group I Paks Promote Skeletal Myoblast Differentiation In Vivo and In Vitro. Mol Cell Biol 2017; 37:MCB.00222-16. [PMID: 27920252 DOI: 10.1128/mcb.00222-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/26/2016] [Indexed: 12/15/2022] Open
Abstract
Skeletal myogenesis is regulated by signal transduction, but the factors and mechanisms involved are not well understood. The group I Paks Pak1 and Pak2 are related protein kinases and direct effectors of Cdc42 and Rac1. Group I Paks are ubiquitously expressed and specifically required for myoblast fusion in Drosophila We report that both Pak1 and Pak2 are activated during mammalian myoblast differentiation. One pathway of activation is initiated by N-cadherin ligation and involves the cadherin coreceptor Cdo with its downstream effector, Cdc42. Individual genetic deletion of Pak1 and Pak2 in mice has no overt effect on skeletal muscle development or regeneration. However, combined muscle-specific deletion of Pak1 and Pak2 results in reduced muscle mass and a higher proportion of myofibers with a smaller cross-sectional area. This phenotype is exacerbated after repair to acute injury. Furthermore, primary myoblasts lacking Pak1 and Pak2 display delayed expression of myogenic differentiation markers and myotube formation. These results identify Pak1 and Pak2 as redundant regulators of myoblast differentiation in vitro and in vivo and as components of the promyogenic Ncad/Cdo/Cdc42 signaling pathway.
Collapse
|
26
|
Krauss RS, Joseph GA, Goel AJ. Keep Your Friends Close: Cell-Cell Contact and Skeletal Myogenesis. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029298. [PMID: 28062562 DOI: 10.1101/cshperspect.a029298] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Development of skeletal muscle is a multistage process that includes lineage commitment of multipotent progenitor cells, differentiation and fusion of myoblasts into multinucleated myofibers, and maturation of myofibers into distinct types. Lineage-specific transcriptional regulation lies at the core of this process, but myogenesis is also regulated by extracellular cues. Some of these cues are initiated by direct cell-cell contact between muscle precursor cells themselves or between muscle precursors and cells of other lineages. Examples of the latter include interaction of migrating neural crest cells with multipotent muscle progenitor cells, muscle interstitial cells with myoblasts, and neurons with myofibers. Among the signaling factors involved are Notch ligands and receptors, cadherins, Ig superfamily members, and Ephrins and Eph receptors. In this article we describe recent progress in this area and highlight open questions raised by the findings.
Collapse
Affiliation(s)
- Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Giselle A Joseph
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Aviva J Goel
- Department of Cell, Developmental, and Regenerative Biology, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
27
|
PKN2 and Cdo interact to activate AKT and promote myoblast differentiation. Cell Death Dis 2016; 7:e2431. [PMID: 27763641 PMCID: PMC5133968 DOI: 10.1038/cddis.2016.296] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 01/01/2023]
Abstract
Skeletal myogenesis is coordinated by multiple signaling pathways that control cell adhesion/migration, survival and differentiation accompanied by muscle-specific gene expression. A cell surface protein Cdo is involved in cell contact-mediated promyogenic signals through activation of p38MAPK and AKT. Protein kinase C-related kinase 2 (PKN2/PRK2) is implicated in regulation of various biological processes, including cell migration, adhesion and death. It has been shown to interact with and inhibit AKT thereby inducing cell death. This led us to investigate the role of PKN2 in skeletal myogenesis and the crosstalk between PKN2 and Cdo. Like Cdo, PKN2 was upregulated in C2C12 myoblasts during differentiation and decreased in cells with Cdo depletion caused by shRNA or cultured on integrin-independent substratum. This decline of PKN2 levels resulted in diminished AKT activation during myoblast differentiation. Consistently, PKN2 overexpression-enhanced C2C12 myoblast differentiation, whereas PKN2-depletion impaired it, without affecting cell survival. PKN2 formed complexes with Cdo, APPL1 and AKT via its C-terminal region and this interaction appeared to be important for induction of AKT activity as well as myoblast differentiation. Furthermore, PKN2-enhanced MyoD-responsive reporter activities by mediating the recruitment of BAF60c and MyoD to the myogenin promoter. Taken together, PKN2 has a critical role in cell adhesion-mediated AKT activation during myoblast differentiation.
Collapse
|
28
|
Leem YE, Jeong HJ, Kim HJ, Koh J, Kang K, Bae GU, Cho H, Kang JS. Cdo Regulates Surface Expression of Kir2.1 K+ Channel in Myoblast Differentiation. PLoS One 2016; 11:e0158707. [PMID: 27380411 PMCID: PMC4933383 DOI: 10.1371/journal.pone.0158707] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 06/21/2016] [Indexed: 01/28/2023] Open
Abstract
A potassium channel Kir2.1-associated membrane hyperpolarization is required for myogenic differentiation. However the molecular regulatory mechanisms modulating Kir2.1 channel activities in early stage of myogenesis are largely unknown. A cell surface protein, Cdo functions as a component of multiprotein cell surface complexes to promote myogenesis. In this study, we report that Cdo forms a complex with Kir2.1 during myogenic differentiation, and is required for the channel activity by enhancing the surface expression of Kir2.1 in the early stage of differentiation. The expression of a constitutively active form of the upstream kinase for p38MAPK, MKK6(EE) can restore Kir2.1 activities in Cdo-depleted C2C12 cells, while the treatment with a p38MAPK inhibitor, SB203580 exhibits a similar effect of Cdo depletion on Kir2.1 surface expression. Furthermore, Cdo-/- primary myoblasts, which display a defective differentiation program, exhibit a defective Kir2.1 activity. Taken together, our results suggest that a promyogenic Cdo signaling is critical for Kir2.1 activities in the induction of myogenic differentiation.
Collapse
Affiliation(s)
- Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Samsung Biomedical Research Institute, Suwon, Republic of Korea
| | - Hyeon-Ju Jeong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Samsung Biomedical Research Institute, Suwon, Republic of Korea
| | - Hyun-Ji Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Samsung Biomedical Research Institute, Suwon, Republic of Korea
| | - Jewoo Koh
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Samsung Biomedical Research Institute, Suwon, Republic of Korea
| | - KyeongJin Kang
- Department of Anatomy, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Samsung Biomedical Research Institute, Suwon, Republic of Korea
| | - Gyu-Un Bae
- Research Center for Cell Fate Control, Sookmyung Women’s University, Seoul, Republic of Korea
| | - Hana Cho
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Samsung Biomedical Research Institute, Suwon, Republic of Korea
- * E-mail: (JSK); (HC)
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
- Samsung Biomedical Research Institute, Suwon, Republic of Korea
- * E-mail: (JSK); (HC)
| |
Collapse
|
29
|
Wang LC, Almazan G. Cdon, a cell surface protein, mediates oligodendrocyte differentiation and myelination. Glia 2016; 64:1021-33. [PMID: 26988125 DOI: 10.1002/glia.22980] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/11/2016] [Indexed: 12/13/2022]
Abstract
During central nervous system development, oligodendrocyte progenitors (OLPs) establish multiple branched processes and axonal contacts to initiate myelination. A complete understanding of the molecular signals implicated in cell surface interaction to initiate myelination/remyelination is currently lacking. The objective of our study was to assess whether Cdon, a cell surface protein that was shown to participate in muscle and neuron cell development, is involved in oligodendrocyte (OLG) differentiation and myelination. Here, we demonstrate that endogenous Cdon protein is expressed in OLPs, increasing in the early differentiation stages and decreasing in mature OLGs. Immunocytochemistry of endogenous Cdon showed localization on both OLG cell membranes and cellular processes exhibiting puncta- or varicosity-like structures. Cdon knockdown with siRNA decreased protein levels by 62% as well as two myelin-specific proteins, MBP and MAG. Conversely, overexpression of full-length rat Cdon increased myelin proteins in OLGs. The complexity of OLGs branching and contact point numbers with axons were also increased in Cdon overexpressing cells growing alone or in coculture with dorsal root ganglion neurons (DRGNs). Furthermore, myelination of DRGNs was decreased when OLPs were transfected with Cdon siRNA. Altogether, our results suggest that Cdon participates in OLG differentiation and myelination, most likely in the initial stages of development.
Collapse
Affiliation(s)
- Li-Chun Wang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6
| | - Guillermina Almazan
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, Quebec, Canada, H3G 1Y6
| |
Collapse
|
30
|
A Shh coreceptor Cdo is required for efficient cardiomyogenesis of pluripotent stem cells. J Mol Cell Cardiol 2016; 93:57-66. [PMID: 26906632 DOI: 10.1016/j.yjmcc.2016.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/13/2016] [Indexed: 11/20/2022]
Abstract
Sonic hedgehog (Shh) signaling plays an important role for early heart development, such as heart looping and cardiomyogenesis of pluripotent stem cells. A multifunctional receptor Cdo functions as a Shh coreceptor together with Boc and Gas1 to activate Shh signaling and these coreceptors seem to play compensatory roles in early heart development. Thus in this study, we examined the role of Cdo in cardiomyogenesis by utilizing an in vitro differentiation of pluripotent stem cells. Here we show that Cdo is required for efficient cardiomyogenesis of pluripotent stem cells by activation of Shh signaling. Cdo is induced concurrently with Shh signaling activation upon induction of cardiomyogenesis of P19 embryonal carcinoma (EC) cells. Cdo-depleted P19 EC and Cdo(-/-) mouse embryonic stem (ES) cells display decreased expression of key cardiac regulators, including Gata4, Nkx2.5 and Mef2c and this decrease coincides with reduced Shh signaling activities. Furthermore Cdo deficiency causes a stark reduction in formation of mature contractile cardiomyocytes. This defect in cardiomyogenesis is overcome by reactivation of Shh signaling at the early specification stage of cardiomyogenesis. The Shh agonist treatment restores differentiation capacities of Cdo-deficient ES cells into contractile cardiomyocytes by recovering both the expression of early cardiac regulators and structural genes such as cardiac troponin T and Connexin 43. Therefore Cdo is required for efficient cardiomyogenesis of pluripotent stem cells and an excellent target to improve the differentiation potential of stem cells for generation of transplantable cells to treat cardiomyopathies.
Collapse
|
31
|
Lee SJ, Go GY, Yoo M, Kim YK, Seo DW, Kang JS, Bae GU. Peroxisome proliferator-activated receptor β/δ (PPARβ/δ) activates promyogenic signaling pathways, thereby promoting myoblast differentiation. Biochem Biophys Res Commun 2016; 470:157-162. [DOI: 10.1016/j.bbrc.2016.01.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 01/04/2016] [Indexed: 11/25/2022]
|
32
|
Martins AF, Xavier Neto J, Azambuja A, Sereno ML, Figueira A, Campos-Junior PH, Rosário MF, Toledo CBB, Silva GAB, Kitten GT, Coutinho LL, Dietrich S, Jorge EC. Repulsive Guidance Molecules a, b and c Are Skeletal Muscle Proteins, and Repulsive Guidance Molecule a Promotes Cellular Hypertrophy and Is Necessary for Myotube Fusion. Cells Tissues Organs 2015; 200:326-38. [PMID: 26397945 DOI: 10.1159/000433491] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2015] [Indexed: 11/19/2022] Open
Abstract
Repulsive guidance molecules (RGMs) compose a family of glycosylphosphatidylinositol (GPI)-anchored axon guidance molecules and perform several functions during neural development. New evidence has suggested possible new roles for these axon guidance molecules during skeletal muscle development, which has not been investigated thus far. In the present study, we show that RGMa, RGMb and RGMc are all induced during skeletal muscle differentiation in vitro. Immunolocalization performed on adult skeletal muscle cells revealed that RGMa, RGMb and RGMc are sarcolemmal proteins. Additionally, RGMa was found to be a sarcoplasmic protein with a surprisingly striated pattern. RGMa colocalization with known sarcoplasmic proteins suggested that this axon guidance molecule is a skeletal muscle sarcoplasmic protein. Western blot analysis revealed two RGMa fragments of 60 and 33 kDa, respectively, in adult skeletal muscle samples. RGMa phenotypes in skeletal muscle cells (C2C12 and primary myoblasts) were also investigated. RGMa overexpression produced hypertrophic cells, whereas RGMa knockdown resulted in the opposite phenotype. RGMa knockdown also blocked myotube formation in both skeletal muscle cell types. Our results are the first to show an axon guidance molecule as a skeletal muscle sarcoplasmic protein and to include RGMa in a system that regulates skeletal muscle cell size and differentiation.
Collapse
Affiliation(s)
- Aline Fagundes Martins
- Departamento de Morfologia, Instituto de Cix00EA;ncias Biolx00F3;gicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Syntaxin 4 regulates the surface localization of a promyogenic receptor Cdo thereby promoting myogenic differentiation. Skelet Muscle 2015; 5:28. [PMID: 26347807 PMCID: PMC4561423 DOI: 10.1186/s13395-015-0052-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/29/2015] [Indexed: 01/05/2023] Open
Abstract
Background Syntaxins are a family of membrane proteins involved in vesicle trafficking, such as synaptic vesicle exocytosis. Syntaxin 4 (Stx4) is expressed highly in skeletal muscle and plays a critical role in insulin-stimulated glucose uptake by promoting translocation of glucose transporter 4 (GLUT4) to the cell surface. A cell surface receptor cell adhesion molecule-related, down-regulated by oncogenes (Cdo) is a component of cell adhesion complexes and promotes myoblast differentiation via activation of key signalings, including p38MAPK and AKT. In this study, we investigate the function of Stx4 in myoblast differentiation and the crosstalk between Stx4 and Cdo in myoblast differentiation. Methods The effects of overexpression or shRNA-based depletion of Stx4 and Cdo genes on C2C12 myoblast differentiation are assessed by Western blotting and immunofluorescence approaches. The interaction between Cdo and Stx4 and the responsible domain mapping are assessed by coimmunoprecipitation or pulldown assays. The effect of Stx4 depletion on cell surface localization of Cdo and GLUT4 in C2C12 myoblasts is assessed by surface biotinylation and Western blotting. Results Overexpression or knockdown of Stx4 enhances or inhibits myogenic differentiation, respectively. Stx4 binds to the cytoplasmic tail of Cdo, and this interaction seems to be critical for induction of p38MAPK activation and myotube formation. Stx4 depletion decreases specifically the cell surface localization of Cdo without changes in surface N-Cadherin levels. Interestingly, Cdo depletion reduces the level of GLUT4 and Stx4 at cell surface. Consistently, overexpression of Cdo in C2C12 myoblasts generally increases glucose uptake, while Cdo depletion reduces it. Conclusions Stx4 promotes myoblast differentiation through interaction with Cdo and stimulation of its surface translocation. Both Cdo and Stx4 are required for GLUT4 translocation to cell surface and glucose uptake in myoblast differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0052-8) contains supplementary material, which is available to authorized users.
Collapse
|
34
|
Cdo suppresses canonical Wnt signalling via interaction with Lrp6 thereby promoting neuronal differentiation. Nat Commun 2014; 5:5455. [PMID: 25406935 DOI: 10.1038/ncomms6455] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 10/02/2014] [Indexed: 01/23/2023] Open
Abstract
Canonical Wnt signalling regulates expansion of neural progenitors and functions as a dorsalizing signal in the developing forebrain. In contrast, the multifunctional co-receptor Cdo promotes neuronal differentiation and is important for the function of the ventralizing signal, Shh. Here we show that Cdo negatively regulates Wnt signalling during neurogenesis. Wnt signalling is enhanced in Cdo-deficient cells, leading to impaired neuronal differentiation. The ectodomains of Cdo and Lrp6 interact via the Ig2 repeat of Cdo and the LDLR repeats of Lrp6, and the Cdo Ig2 repeat is necessary for Cdo-dependent Wnt inhibition. Furthermore, the Cdo-deficient dorsal forebrain displays stronger Wnt signalling activity, increased cell proliferation and enhanced expression of the dorsal markers and Wnt targets, Pax6, Gli3, Axin2. Therefore, in addition to promoting ventral central nervous system cell fates with Shh, Cdo promotes neuronal differentiation by suppression of Wnt signalling and provides a direct link between two major dorsoventral morphogenetic signalling pathways.
Collapse
|
35
|
Yi P, Chew LL, Zhang Z, Ren H, Wang F, Cong X, Zheng L, Luo Y, Ouyang H, Low BC, Zhou YT. KIF5B transports BNIP-2 to regulate p38 mitogen-activated protein kinase activation and myoblast differentiation. Mol Biol Cell 2014; 26:29-42. [PMID: 25378581 PMCID: PMC4279227 DOI: 10.1091/mbc.e14-03-0797] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cdo bridges scaffold proteins BNIP-2 and JLP to activate p38MAPK during myoblast differentiation. KIF5B is a novel interacting partner of BNIP-2 and promotes myogenic differentiation. KIF5B-dependent transport of BNIP-2 is essential for its promyogenic effects. The Cdo-p38MAPK (p38 mitogen-activated protein kinase) signaling pathway plays important roles in regulating skeletal myogenesis. During myogenic differentiation, the cell surface receptor Cdo bridges scaffold proteins BNIP-2 and JLP and activates p38MAPK, but the spatial-temporal regulation of this process is largely unknown. We here report that KIF5B, the heavy chain of kinesin-1 motor, is a novel interacting partner of BNIP-2. Coimmunoprecipitation and far-Western study revealed that BNIP-2 directly interacted with the motor and tail domains of KIF5B via its BCH domain. By using a range of organelle markers and live microscopy, we determined the endosomal localization of BNIP-2 and revealed the microtubule-dependent anterograde transport of BNIP-2 in C2C12 cells. The anterograde transport of BNIP-2 was disrupted by a dominant-negative mutant of KIF5B. In addition, knockdown of KIF5B causes aberrant aggregation of BNIP-2, confirming that KIF5B is critical for the anterograde transport of BNIP-2 in cells. Gain- and loss-of-function experiments further showed that KIF5B modulates p38MAPK activity and in turn promotes myogenic differentiation. Of importance, the KIF5B-dependent anterograde transport of BNIP-2 is critical for its promyogenic effects. Our data reveal a novel role of KIF5B in the spatial regulation of Cdo–BNIP-2–p38MAPK signaling and disclose a previously unappreciated linkage between the intracellular transporting system and myogenesis regulation.
Collapse
Affiliation(s)
- Peng Yi
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Li Li Chew
- Department of Biological Sciences and Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Ziwang Zhang
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hao Ren
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feiya Wang
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoxia Cong
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Liling Zheng
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and
| | - Yan Luo
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and
| | - Hongwei Ouyang
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Boon Chuan Low
- Department of Biological Sciences and Mechanobiology Institute, National University of Singapore, 117411 Singapore
| | - Yi Ting Zhou
- Center for Stem Cell and Tissue Engineering, Department of Biochemistry and Molecular Biology, and Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
36
|
Abstract
Muscle fibers form as a result of myoblast fusion, yet the cell surface receptors regulating this process are unknown in vertebrates. In Drosophila, myoblast fusion involves the activation of the Rac pathway by the guanine nucleotide exchange factor Myoblast City and its scaffolding protein ELMO, downstream of cell-surface cell-adhesion receptors. We previously showed that the mammalian ortholog of Myoblast City, DOCK1, functions in an evolutionarily conserved manner to promote myoblast fusion in mice. In search for regulators of myoblast fusion, we identified the G-protein coupled receptor brain-specific angiogenesis inhibitor (BAI3) as a cell surface protein that interacts with ELMO. In cultured cells, BAI3 or ELMO1/2 loss of function severely impaired myoblast fusion without affecting differentiation and cannot be rescued by reexpression of BAI3 mutants deficient in ELMO binding. The related BAI protein family member, BAI1, is functionally distinct from BAI3, because it cannot rescue the myoblast fusion defects caused by the loss of BAI3 function. Finally, embryonic muscle precursor expression of a BAI3 mutant unable to bind ELMO was sufficient to block myoblast fusion in vivo. Collectively, our findings provide a role for BAI3 in the relay of extracellular fusion signals to their intracellular effectors, identifying it as an essential transmembrane protein for embryonic vertebrate myoblast fusion.
Collapse
|
37
|
Trajkovski M, Lodish H. MicroRNA networks regulate development of brown adipocytes. Trends Endocrinol Metab 2013; 24:442-50. [PMID: 23809233 PMCID: PMC3979327 DOI: 10.1016/j.tem.2013.05.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/10/2013] [Accepted: 05/13/2013] [Indexed: 12/22/2022]
Abstract
Brown adipose tissue (BAT) is specialized for heat generation and energy expenditure as a defense against cold and obesity; in both humans and mice increased amounts of BAT are associated with a lean phenotype and resistance to development of the metabolic syndrome and its complications. Here we summarize recent research showing that several BAT-expressed microRNAs (miRNAs) play important roles in regulating differentiation and metabolism of brown and beige adipocytes; we discuss the key mRNA targets downregulated by these miRNAs and show how these miRNAs affect directly or indirectly transcription factors important for BAT development. We suggest that these miRNAs could be part of novel therapeutics to increase BAT in humans.
Collapse
Affiliation(s)
- Mirko Trajkovski
- University College London (UCL), Division of Biosciences, School of Life and Medical Sciences, Institute of Structural and Molecular Biology, Darwin building, Gower street, London WC1E 6BT, UK.
| | | |
Collapse
|
38
|
Romer AI, Singh J, Rattan S, Krauss RS. Smooth muscle fascicular reorientation is required for esophageal morphogenesis and dependent on Cdo. ACTA ACUST UNITED AC 2013; 201:309-23. [PMID: 23569214 PMCID: PMC3628509 DOI: 10.1083/jcb.201301005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cdo-deficient mice have defects in smooth muscle fascicular reorientation during esophageal morphogenesis, resulting in structural and functional defects including an aberrantly proximal skeletal–smooth muscle boundary and achalasia. Postnatal maturation of esophageal musculature involves proximal-to-distal replacement of smooth muscle with skeletal muscle by elusive mechanisms. We report that this process is impaired in mice lacking the cell surface receptor Cdo and identify the underlying developmental mechanism. A myogenic transition zone containing proliferative skeletal muscle precursor cells migrated in a proximal–distal direction, leaving differentiated myofibers in its wake. Distal to the transition zone, smooth muscle fascicles underwent a morphogenetic process whereby they changed their orientation relative to each other and to the lumen. Consequently, a path was cleared for the transition zone, and smooth muscle ultimately occupied only the distal-most esophagus; there was no loss of smooth muscle. Cdo−/− mice were specifically defective in fascicular reorientation, resulting in an aberrantly proximal skeletal–smooth muscle boundary. Furthermore, Cdo−/− mice displayed megaesophagus and achalasia, and their lower esophageal sphincter was resistant to nitric oxide–induced relaxation, suggesting a developmental linkage between patterning and sphincter function. Collectively, these results illuminate mechanisms of esophageal morphogenesis and motility disorders.
Collapse
Affiliation(s)
- Anthony I Romer
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | |
Collapse
|
39
|
Zhang H, Chen Y, Chen Y. Noise propagation in gene regulation networks involving interlinked positive and negative feedback loops. PLoS One 2012; 7:e51840. [PMID: 23284787 PMCID: PMC3527455 DOI: 10.1371/journal.pone.0051840] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 11/13/2012] [Indexed: 01/30/2023] Open
Abstract
It is well known that noise is inevitable in gene regulatory networks due to the low-copy numbers of molecules and local environmental fluctuations. The prediction of noise effects is a key issue in ensuring reliable transmission of information. Interlinked positive and negative feedback loops are essential signal transduction motifs in biological networks. Positive feedback loops are generally believed to induce a switch-like behavior, whereas negative feedback loops are thought to suppress noise effects. Here, by using the signal sensitivity (susceptibility) and noise amplification to quantify noise propagation, we analyze an abstract model of the Myc/E2F/MiR-17-92 network that is composed of a coupling between the E2F/Myc positive feedback loop and the E2F/Myc/miR-17-92 negative feedback loop. The role of the feedback loop on noise effects is found to depend on the dynamic properties of the system. When the system is in monostability or bistability with high protein concentrations, noise is consistently suppressed. However, the negative feedback loop reduces this suppression ability (or improves the noise propagation) and enhances signal sensitivity. In the case of excitability, bistability, or monostability, noise is enhanced at low protein concentrations. The negative feedback loop reduces this noise enhancement as well as the signal sensitivity. In all cases, the positive feedback loop acts contrary to the negative feedback loop. We also found that increasing the time scale of the protein module or decreasing the noise autocorrelation time can enhance noise suppression; however, the systems sensitivity remains unchanged. Taken together, our results suggest that the negative/positive feedback mechanisms in coupled feedback loop dynamically buffer noise effects rather than only suppressing or amplifying the noise.
Collapse
Affiliation(s)
- Hui Zhang
- Institute of Theoretical Physics, Lanzhou University, Lanzhou, China
| | - Yueling Chen
- Institute of Theoretical Physics, Lanzhou University, Lanzhou, China
- Department of Physics, Gansu College of Traditional Chinese Medicine, Lanzhou, China
| | - Yong Chen
- Institute of Theoretical Physics, Lanzhou University, Lanzhou, China
- Department of Mathematics, Kings College London, London, United Kingdom
| |
Collapse
|
40
|
Tran P, Ho SM, Kim BG, Vuong TA, Leem YE, Bae GU, Kang JS. TGF-β-activated kinase 1 (TAK1) and apoptosis signal-regulating kinase 1 (ASK1) interact with the promyogenic receptor Cdo to promote myogenic differentiation via activation of p38MAPK pathway. J Biol Chem 2012; 287:11602-15. [PMID: 22337877 DOI: 10.1074/jbc.m112.351601] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
p38MAPK plays an essential role in the transition of myoblasts to differentiated myotubes through the activation of MyoD family transcription factors. A promyogenic cell surface molecule, Cdo, promotes myogenic differentiation mainly through activation of the p38MAPK pathway. Two MAP3Ks, TAK1 and ASK1, can activate p38MAPK via MKK6 in various cell systems. Moreover TAK1 has been shown to promote myogenic differentiation via p38MAPK activation. In this study, we hypothesized that TAK1 and ASK1 might function as MAP3Ks in Cdo-mediated p38MAPK activation during myoblast differentiation. Both ASK1 and TAK1 were expressed in myoblasts and interacted with the cytoplasmic tail of Cdo and a scaffold protein, JLP. The depletion of TAK1 or ASK1 in C2C12 cells decreased myoblast differentiation, whereas overexpression of TAK1 or ASK1 in C2C12 cells enhanced myotube formation. In agreement with this, overexpression of ASK1 or TAK1 resulted in enhanced p38MAPK activation, and their knockdown inhibited p38MAPK in C2C12 cells. Overexpression of TAK1 or ASK1 in Cdo(-/-) myoblasts and Cdo-depleted C2C12 cells restored p38MAPK activation as well as myotube formation. Furthermore, ASK1 and TAK1 compensated for each other in p38MAPK activation and myoblast differentiation. Taken together, these findings suggest that ASK1 and TAK1 function as MAP3Ks in Cdo-mediated p38MAPK activation to promote myogenic differentiation.
Collapse
Affiliation(s)
- Phong Tran
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Cdon and Boc: Two transmembrane proteins implicated in cell-cell communication. Int J Biochem Cell Biol 2012; 44:698-702. [PMID: 22326621 DOI: 10.1016/j.biocel.2012.01.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 01/20/2012] [Accepted: 01/27/2012] [Indexed: 11/23/2022]
Abstract
Cdon and Boc, and their Drosophila homologues Ihog and Boi, are evolutionary conserved transmembrane glycoproteins belonging to a subgroup of the Immunoglobulin superfamily of cell adhesion molecules (CAMs). Initially isolated in vertebrates as CAMs that link cadherin function with MAPK signaling in myoblast differentiation, they have thereafter been shown to act as essential receptors for the Hedgehog (Hh) family of secreted proteins. They associate with both ligand and other Hh receptor components, including Ptch and Gas1, thus forming homo- and heteromeric complexes. In Drosophila, they are also involved in ligand processing and release from Hh producing cells. Cdon/Boc and Ihog/Boi can substitute one another and play redundant functions is some contexts. In addition, Boc, but not Cdon, mediates axon guidance information provided by Hh in specific neuronal populations, whereas mutations in the CDON cause holoprosencephaly, a human congenital anomaly defined by forebrain midline defects prominently associated with diminished Hh pathway activity.
Collapse
|
42
|
Lee HJ, Bae GU, Leem YE, Choi HK, Kang TM, Cho H, Kim ST, Kang JS. Phosphorylation of Stim1 at serine 575 via netrin-2/Cdo-activated ERK1/2 is critical for the promyogenic function of Stim1. Mol Biol Cell 2012; 23:1376-87. [PMID: 22298426 PMCID: PMC3315807 DOI: 10.1091/mbc.e11-07-0634] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The promyogenic cell surface molecule Cdo is required for activation of extracellular signal-regulated kinase (ERK) and nuclear factor of activated T cells c3 (NFATc3) induced by netrin-2 in myogenic differentiation. However, the molecular mechanism leading to NFATc3 activation is unknown. Stromal interaction molecule 1 (Stim1), an internal calcium sensor of the endoplasmic reticulum store, promotes myogenesis via activation of NFATc3. In this study we investigated the functional interaction between Cdo and Stim1 in myogenic differentiation. Overexpression and depletion of Stim1 enhanced or decreased myotube formation, respectively. Of interest, Stim1 protein levels were decreased in Cdo-deficient perinatal hindlimb muscles or primary myoblasts; this correlates with defective NFATc3 activation in Cdo(-/-) myoblasts upon differentiation. Forced activation of NFATc3 by overexpression of calcineurin restored differentiation of Cdo-depleted C2C12 myoblasts. Furthermore, Cdo and Stim1 formed a complex in 293T cells or in differentiating C2C12 myoblasts. The netrin-2-mediated NFATc3 activation was coincident with robust interactions between Cdo and Stim1 in myoblasts and the ERK-mediated Stim1 phosphorylation at serine 575. The serine 575 phosphorylation was enhanced in C2C12 cells upon differentiation, and the alanine substitution of serine 575 failed to restore differentiation of Stim1-depleted myoblasts. Taken together, the results indicate that cell adhesion signaling triggered by netrin-2/Cdo induces Stim1 phosphorylation at serine 575 by ERK, which promotes myoblast differentiation.
Collapse
Affiliation(s)
- Hye-Jin Lee
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Mancini A, Sirabella D, Zhang W, Yamazaki H, Shirao T, Krauss RS. Regulation of myotube formation by the actin-binding factor drebrin. Skelet Muscle 2011; 1:36. [PMID: 22152295 PMCID: PMC3251523 DOI: 10.1186/2044-5040-1-36] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 12/08/2011] [Indexed: 11/15/2022] Open
Abstract
Background Myogenic differentiation involves cell-cycle arrest, activation of the muscle-specific transcriptome, and elongation, alignment and fusion of myoblasts into multinucleated myotubes. This process is controlled by promyogenic transcription factors and regulated by signaling pathways in response to extracellular cues. The p38 mitogen-activated protein kinase (p38 MAPK) pathway promotes the activity of several such transcription factors, including MyoD and MEF2, thereby controlling the muscle-specific transcription program. However, few p38-regulated genes that play a role in the regulation of myogenesis have been identified. Methods RNA interference (RNAi), chemical inhibition and immunofluorescence approaches were used to assess the role of drebrin in differentiation of primary mouse myoblasts and C2C12 cells. Results In a search for p38-regulated genes that promote myogenic differentiation, we identified Dbn1, which encodes the actin-binding protein drebrin. Drebrin is an F-actin side-binding protein that remodels actin to facilitate the change of filopodia into dendritic spines during synaptogenesis in developing neurons. Dbn1 mRNA and protein are induced during differentiation of primary mouse and C2C12 myoblasts, and induction is substantially reduced by the p38 MAPK inhibitor SB203580. Primary myoblasts and C2C12 cells depleted of drebrin by RNAi display reduced levels of myogenin and myosin heavy chain and form multinucleated myotubes very inefficiently. Treatment of myoblasts with BTP2, a small-molecule inhibitor of drebrin, produces a phenotype similar to that produced by knockdown of drebrin, and the inhibitory effects of BTP2 are rescued by expression of a mutant form of drebrin that is unable to bind BTP2. Drebrin in myoblasts is enriched in cellular projections and cell cortices and at regions of cell-cell contact, all sites where F-actin, too, was concentrated. Conclusions Our findings reveal that Dbn1 expression is a target of p38 MAPK signaling during myogenesis and that drebrin promotes myoblast differentiation.
Collapse
Affiliation(s)
- Annalisa Mancini
- Department of Developmental and Regenerative Biology, Mount Sinai School of Medicine, One Gustave L, Levy Place, New York, NY 10029, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Leem YE, Han JW, Lee HJ, Ha HL, Kwon YL, Ho SM, Kim BG, Tran P, Bae GU, Kang JS. Gas1 cooperates with Cdo and promotes myogenic differentiation via activation of p38MAPK. Cell Signal 2011; 23:2021-9. [DOI: 10.1016/j.cellsig.2011.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 07/18/2011] [Indexed: 12/01/2022]
|
45
|
Mutations in CDON, encoding a hedgehog receptor, result in holoprosencephaly and defective interactions with other hedgehog receptors. Am J Hum Genet 2011; 89:231-40. [PMID: 21802063 DOI: 10.1016/j.ajhg.2011.07.001] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Revised: 06/09/2011] [Accepted: 07/05/2011] [Indexed: 11/20/2022] Open
Abstract
Holoprosencephaly (HPE), a common human congenital anomaly defined by a failure to delineate the midline of the forebrain and/or midface, is associated with diminished Sonic hedgehog (SHH)-pathway activity in development of these structures. SHH signaling is regulated by a network of ligand-binding factors, including the primary receptor PTCH1 and the putative coreceptors, CDON (also called CDO), BOC, and GAS1. Although binding of SHH to these receptors promotes pathway activity, it is not known whether interactions between these receptors are important. We report here identification of missense CDON mutations in human HPE. These mutations diminish CDON's ability to support SHH-dependent gene expression in cell-based signaling assays. The mutations occur outside the SHH-binding domain of CDON, and the encoded variant CDON proteins do not display defects in binding to SHH. In contrast, wild-type CDON associates with PTCH1 and GAS1, but the variants do so inefficiently, in a manner that parallels their activity in cell-based assays. Our findings argue that CDON must associate with both ligand and other hedgehog-receptor components, particularly PTCH1, for signaling to occur and that disruption of the latter interactions is a mechanism of HPE.
Collapse
|
46
|
Molecular and cellular mechanisms of mammalian cell fusion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 713:33-64. [PMID: 21432013 DOI: 10.1007/978-94-007-0763-4_4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The fusion of one cell with another occurs in development, injury and disease. Despite the diversity of fusion events, five steps in sequence appear common. These steps include programming fusion-competent status, chemotaxis, membrane adhesion, membrane fusion, and post-fusion resetting. Recent advances in the field start to reveal the molecules involved in each step. This review focuses on some key molecules and cellular events of cell fusion in mammals. Increasing evidence demonstrates that membrane lipid rafts, adhesion proteins and actin rearrangement are critical in the final step of membrane fusion. Here we propose a new model for the formation and expansion of membrane fusion pores based on recent observations on myotube formation. In this model, membrane lipid rafts first recruit adhesion molecules and align with opposing membranes, with the help of a cortical actin "wall" as a rigid supportive platform. Second, the membrane adhesion proteins interact with each other and trigger actin rearrangement, which leads to rapid dispersion of lipid rafts and flow of a highly fluidic phospholipid bilayer into the site. Finally, the opposing phospholipid bilayers are then pushed into direct contact leading to the formation of fusion pores by the force generated through actin polymerization. The actin polymerization generated force also drives the expansion of the fusion pores. However, several key questions about the process of cell fusion still remain to be explored. The understanding of the mechanisms of cell fusion may provide new opportunities in correcting development disorders or regenerating damaged tissues by inhibiting or promoting molecular events associated with fusion.
Collapse
|
47
|
Sun L, Xie H, Mori MA, Alexander R, Yuan B, Hattangadi SM, Liu Q, Kahn CR, Lodish HF. Mir193b-365 is essential for brown fat differentiation. Nat Cell Biol 2011; 13:958-65. [PMID: 21743466 PMCID: PMC3149720 DOI: 10.1038/ncb2286] [Citation(s) in RCA: 244] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 05/27/2011] [Indexed: 01/14/2023]
Abstract
Mammals have two principal types of fat. White adipose tissue primarily serves to store extra energy as triglycerides, whereas brown adipose tissue is specialized to burn lipids for heat generation and energy expenditure as a defence against cold and obesity. Recent studies have demonstrated that brown adipocytes arise in vivo from a Myf5-positive, myoblastic progenitor by the action of Prdm16 (PR domain containing 16). Here, we identified a brown-fat-enriched miRNA cluster, MiR-193b-365, as a key regulator of brown fat development. Blocking miR-193b and/or miR-365 in primary brown preadipocytes markedly impaired brown adipocyte adipogenesis by enhancing Runx1t1 (runt-related transcription factor 1; translocated to, 1) expression, whereas myogenic markers were significantly induced. Forced expression of Mir193b and/or Mir365 in C2C12 myoblasts blocked the entire programme of myogenesis, and, in adipogenic conditions, miR-193b induced myoblasts to differentiate into brown adipocytes. Mir193b-365 was upregulated by Prdm16 partially through Pparα. Our results demonstrate that Mir193b-365 serves as an essential regulator for brown fat differentiation, in part by repressing myogenesis.
Collapse
Affiliation(s)
- Lei Sun
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Huangming Xie
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
- Computation and Systems Biology, Singapore-MIT Alliance, National University of Singapore, 4 Engineering Drive 3, Singapore 117576
| | - Marcelo A Mori
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryan Alexander
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bingbing Yuan
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Shilpa M. Hattangadi
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
- Department of Hematology, Boston Children’s Hospital, Boston, MA 02115
| | - Qingqing Liu
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Harvey F. Lodish
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Cambridge, MA 02142, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
48
|
Han JW, Lee HJ, Bae GU, Kang JS. Promyogenic function of Integrin/FAK signaling is mediated by Cdo, Cdc42 and MyoD. Cell Signal 2011; 23:1162-9. [DOI: 10.1016/j.cellsig.2011.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Accepted: 03/03/2011] [Indexed: 12/11/2022]
|
49
|
Secretome Analysis of Skeletal Myogenesis Using SILAC and Shotgun Proteomics. INTERNATIONAL JOURNAL OF PROTEOMICS 2011; 2011:329467. [PMID: 22084683 PMCID: PMC3200090 DOI: 10.1155/2011/329467] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 01/26/2011] [Indexed: 12/18/2022]
Abstract
Myogenesis, the formation of skeletal muscle, is a multistep event that commences with myoblast proliferation, followed by cell-cycle arrest, and finally the formation of multinucleated myotubes via fusion of mononucleated myoblasts. Each step is orchestrated by well-documented intracellular factors, such as cytoplasmic signalling molecules and nuclear transcription factors. Regardless, the key step in getting a more comprehensive understanding of the regulation of myogenesis is to explore the extracellular factors that are capable of eliciting the downstream intracellular factors. This could further provide valuable insight into the acute cellular response to extrinsic cues in maintaining normal muscle development. In this paper, we survey the intracellular factors that respond to extracellular cues that are responsible for the cascades of events during myogenesis: myoblast proliferation, cell-cycle arrest of myoblasts, and differentiation of myoblasts into myotubes. This focus on extracellular perspective of muscle development illustrates our mass spectrometry-based proteomic approaches to identify differentially expressed secreted factors during skeletal myogenesis.
Collapse
|
50
|
Abstract
Satellite cells (SCs) are the main source of adult skeletal muscle stem cells responsible for muscle growth and regeneration. By interpreting extracellular cues, developmental regulators control quiescence, proliferation, and differentiation of SCs by influencing coordinate gene expression. The scope of this review is limited to the description and discussion of protein complexes that introduce and decode heritable histone and chromatin modifications and how these modifications are relevant for SC biology.
Collapse
Affiliation(s)
- Vittorio Sartorelli
- Laboratory of Muscle Stem Cell and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|