1
|
Chi C, Song K. Cellular reprogramming of fibroblasts in heart regeneration. J Mol Cell Cardiol 2023; 180:84-93. [PMID: 36965699 PMCID: PMC10347886 DOI: 10.1016/j.yjmcc.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Myocardial infarction causes the loss of cardiomyocytes and the formation of cardiac fibrosis due to the activation of cardiac fibroblasts, leading to cardiac dysfunction and heart failure. Unfortunately, current therapeutic interventions can only slow the disease progression. Furthermore, they cannot fully restore cardiac function, likely because the adult human heart lacks sufficient capacity to regenerate cardiomyocytes. Therefore, intensive efforts have focused on developing therapeutics to regenerate the damaged heart. Several strategies have been intensively investigated, including stimulation of cardiomyocyte proliferation, transplantation of stem cell-derived cardiomyocytes, and conversion of fibroblasts into cardiac cells. Resident cardiac fibroblasts are critical in the maintenance of the structure and contractility of the heart. Fibroblast plasticity makes this type of cells be reprogrammed into many cell types, including but not limited to induced pluripotent stem cells, induced cardiac progenitor cells, and induced cardiomyocytes. Fibroblasts have become a therapeutic target due to their critical roles in cardiac pathogenesis. This review summarizes the reprogramming of fibroblasts into induced pluripotent stem cell-derived cardiomyocytes, induced cardiac progenitor cells, and induced cardiomyocytes to repair a damaged heart, outlines recent findings in utilizing fibroblast-derived cells for heart regeneration, and discusses the limitations and challenges.
Collapse
Affiliation(s)
- Congwu Chi
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
2
|
Canté-Barrett K, Meijer MT, Cordo' V, Hagelaar R, Yang W, Yu J, Smits WK, Nulle ME, Jansen JP, Pieters R, Yang JJ, Haigh JJ, Goossens S, Meijerink JP. MEF2C opposes Notch in lymphoid lineage decision and drives leukemia in the thymus. JCI Insight 2022; 7:150363. [PMID: 35536646 PMCID: PMC9310523 DOI: 10.1172/jci.insight.150363] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/04/2022] [Indexed: 11/25/2022] Open
Abstract
Rearrangements that drive ectopic MEF2C expression have recurrently been found in patients with human early thymocyte progenitor acute lymphoblastic leukemia (ETP-ALL). Here, we show high levels of MEF2C expression in patients with ETP-ALL. Using both in vivo and in vitro models of ETP-ALL, we demonstrate that elevated MEF2C expression blocks NOTCH-induced T cell differentiation while promoting a B-lineage program. MEF2C activates a B cell transcriptional program in addition to RUNX1, GATA3, and LMO2; upregulates the IL-7R; and boosts cell survival by upregulation of BCL2. MEF2C and the Notch pathway, therefore, demarcate opposite regulators of B- or T-lineage choices, respectively. Enforced MEF2C expression in mouse or human progenitor cells effectively blocks early T cell differentiation and promotes the development of biphenotypic lymphoid tumors that coexpress CD3 and CD19, resembling human mixed phenotype acute leukemia. Salt-inducible kinase (SIK) inhibitors impair MEF2C activity and alleviate the T cell developmental block. Importantly, this sensitizes cells to prednisolone treatment. Therefore, SIK-inhibiting compounds such as dasatinib are potentially valuable additions to standard chemotherapy for human ETP-ALL.
Collapse
Affiliation(s)
| | - Mariska T Meijer
- Princess Máxima Center for pediatric oncology, Utrecht, Netherlands
| | - Valentina Cordo'
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Rico Hagelaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Wentao Yang
- Department of Pharmaceutical Sciences, St. Jude Childen's Research Hospital, Memphis, United States of America
| | - Jiyang Yu
- Computational Biology Department, St. Jude Childen's Research Hospital, Memphis, United States of America
| | - Willem K Smits
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Marloes E Nulle
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Joris P Jansen
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Rob Pieters
- Pieters Group, Princess Máxima Center for pediatric oncology, Utrecht, Netherlands
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, United States of America
| | - Jody J Haigh
- Research Institute of Oncology and Hematology, University of Manitoba, Manitoba, Canada
| | - Steven Goossens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jules Pp Meijerink
- Meijerink Group, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| |
Collapse
|
3
|
Ma X, La Y, Bao P, Chu M, Guo X, Wu X, Pei J, Ding X, Liang C, Yan P. Regulatory Role of N6-Methyladenosine in Longissimus Dorsi Development in Yak. Front Vet Sci 2022; 9:757115. [PMID: 35498742 PMCID: PMC9043854 DOI: 10.3389/fvets.2022.757115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
N6-methyladenine (m6A) RNA undergoes epigenetic modification, which is the most extensive intermediate chemical modification in mRNA. Although this modification occurs in all living organisms, it is the most widely studied among mammals. However, to date, no study has investigated the m6A transcriptome-wide map of yak and its potential biological functions in muscle development. In this study, the differences of m6A methylation and gene expression in yak muscle development belonging to three age groups, namely 3 years (group A), 6 months (group M), and 90-day-old fetuses (group E), were determined by using methylated RNA immunoprecipitation sequencing (MeRIP-Seq) and RNA sequencing (RNA-Seq). In these three groups, a total of 6,278 (A), 9,298 (E), and 9,584 (M) m6A peaks were identified, with average densities between 1.02 and 2.01. m6A peaks were mostly enriched in the stop codon, 3′ untranslated region (UTR) region, and inner long exon region with consensus motifs of UGACA. In all the three stages, the m6A peak enrichment level was significantly negatively correlated with mRNA abundance (Pearson's correlation coefficient r = −0.22 to −0.32, p < 10−16). The functional enrichment of genes consistently modified by m6A methylation, particularly those genes that regulate cell differentiation as well as muscle growth and development, was observed at all three stages. Moreover, m6A abundance was negatively associated with gene expression levels, indicating that m6A might play a vital role in modulating gene expression during yak muscle development. This comprehensive map thus provides a solid foundation for determining the potential functional role of m6A RNA modification in yak muscle growth.
Collapse
Affiliation(s)
- Xiaoming Ma
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yongfu La
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengjia Bao
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Min Chu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xian Guo
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiaoyun Wu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jie Pei
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xuezhi Ding
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Chunnian Liang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
- *Correspondence: Chunnian Liang
| | - Ping Yan
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Ping Yan
| |
Collapse
|
4
|
Aránega AE, Lozano-Velasco E, Rodriguez-Outeiriño L, Ramírez de Acuña F, Franco D, Hernández-Torres F. MiRNAs and Muscle Regeneration: Therapeutic Targets in Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22084236. [PMID: 33921834 PMCID: PMC8072594 DOI: 10.3390/ijms22084236] [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: 04/05/2021] [Accepted: 04/15/2021] [Indexed: 12/17/2022] Open
Abstract
microRNAs (miRNAs) are small non-coding RNAs required for the post-transcriptional control of gene expression. MicroRNAs play a critical role in modulating muscle regeneration and stem cell behavior. Muscle regeneration is affected in muscular dystrophies, and a critical point for the development of effective strategies for treating muscle disorders is optimizing approaches to target muscle stem cells in order to increase the ability to regenerate lost tissue. Within this framework, miRNAs are emerging as implicated in muscle stem cell response in neuromuscular disorders and new methodologies to regulate the expression of key microRNAs are coming up. In this review, we summarize recent advances highlighting the potential of miRNAs to be used in conjunction with gene replacement therapies, in order to improve muscle regeneration in the context of Duchenne Muscular Dystrophy (DMD).
Collapse
Affiliation(s)
- Amelia Eva Aránega
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
- Correspondence:
| | - Estefanía Lozano-Velasco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Lara Rodriguez-Outeiriño
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Felicitas Ramírez de Acuña
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
| | - Francisco Hernández-Torres
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, Paraje Las Lagunillas s/n, 23009 Jaen, Spain; (E.L.-V.); (L.R.-O.); (F.R.d.A.); (D.F.); (F.H.-T.)
- Medina Foundation, Technology Park of Health Sciences, Av. del Conocimiento 34, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, Avda. de la Investigación 11, 18016 Granada, Spain
| |
Collapse
|
5
|
Riching AS, Song K. Cardiac Regeneration: New Insights Into the Frontier of Ischemic Heart Failure Therapy. Front Bioeng Biotechnol 2021; 8:637538. [PMID: 33585427 PMCID: PMC7873479 DOI: 10.3389/fbioe.2020.637538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/29/2020] [Indexed: 12/17/2022] Open
Abstract
Ischemic heart disease is the leading cause of morbidity and mortality in the world. While pharmacological and surgical interventions developed in the late twentieth century drastically improved patient outcomes, mortality rates over the last two decades have begun to plateau. Following ischemic injury, pathological remodeling leads to cardiomyocyte loss and fibrosis leading to impaired heart function. Cardiomyocyte turnover rate in the adult heart is limited, and no clinical therapies currently exist to regenerate cardiomyocytes lost following ischemic injury. In this review, we summarize the progress of therapeutic strategies including revascularization and cell-based interventions to regenerate the heart: transiently inducing cardiomyocyte proliferation and direct reprogramming of fibroblasts into cardiomyocytes. Moreover, we highlight recent mechanistic insights governing these strategies to promote heart regeneration and identify current challenges in translating these approaches to human patients.
Collapse
Affiliation(s)
- Andrew S. Riching
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Pharmacology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
6
|
Yu A, Zheng Y, Gong Y, Yu R, Yang Z, Chen X. Adiponectin promotes myogenic differentiation via a Mef2C-AdipoR1 positive feedback loop. Gene 2020; 771:145380. [PMID: 33359123 DOI: 10.1016/j.gene.2020.145380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/17/2020] [Accepted: 12/15/2020] [Indexed: 11/16/2022]
Abstract
Adiponectin is an important hormone that regulates systemic metabolism, and it has been reported that globular adiponectin promotes myogenic differentiation. However, the mechanisms by which adiponectin promotes myogenic differentiation is not fully understood. In the present study, we show that adiponectin and its receptor 1 are significantly up-regulated during myogenic differentiation and that adiponectin increased the expression level of a core myogenic regulator, Mef2C, which is required for the effects of adiponectin on promoting myogenic differentiation. A transcriptional inhibitor of Mef2C, HDAC9, was down-regulated by adiponectin. In turn, Mef2C overexpression up-regulates adiponectin and its receptor, AdipoR1, to increase myogenic differentiation. We showed that mechanistically, Mef2C directly binds to AdipoR1 promoter to transcriptionally up-regulate AdipoR1 expression, which is required for the effects of Mef2C overexpression on myogenic differentiation. Thus, adiponectin/AdipoR1 and Mef2c form a positive feedback loop to promote myogenic differentiation.
Collapse
Affiliation(s)
- An Yu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yu Zheng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yuqing Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ruofan Yu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zaiqing Yang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China.
| |
Collapse
|
7
|
Sucharov J, Ray K, Brooks EP, Nichols JT. Selective breeding modifies mef2ca mutant incomplete penetrance by tuning the opposing Notch pathway. PLoS Genet 2019; 15:e1008507. [PMID: 31790396 PMCID: PMC6907857 DOI: 10.1371/journal.pgen.1008507] [Citation(s) in RCA: 10] [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: 06/28/2019] [Revised: 12/12/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
Deleterious genetic mutations allow developmental biologists to understand how genes control development. However, not all loss of function genetic mutants develop phenotypic changes. Many deleterious mutations only produce a phenotype in a subset of mutant individuals, a phenomenon known as incomplete penetrance. Incomplete penetrance can confound analyses of gene function and our understanding of this widespread phenomenon remains inadequate. To better understand what controls penetrance, we capitalized on the zebrafish mef2ca mutant which produces craniofacial phenotypes with variable penetrance. Starting with a characterized mef2ca loss of function mutant allele, we used classical selective breeding methods to generate zebrafish strains in which mutant-associated phenotypes consistently appear with low or high penetrance. Strikingly, our selective breeding for low penetrance converted the mef2ca mutant allele behavior from homozygous lethal to homozygous viable. Meanwhile, selective breeding for high penetrance converted the mef2ca mutant allele from fully recessive to partially dominant. Comparing the selectively-bred low- and high-penetrance strains revealed that the strains initially respond similarly to the mutation, but then gene expression differences between strains emerge during development. Thus, altered temporal genetic circuitry can manifest through selective pressure to modify mutant penetrance. Specifically, we demonstrate differences in Notch signaling between strains, and further show that experimental manipulation of the Notch pathway phenocopies penetrance changes occurring through selective breeding. This study provides evidence that penetrance is inherited as a liability-threshold trait. Our finding that vertebrate animals can overcome a deleterious mutation by tuning genetic circuitry complements other reported mechanisms of overcoming deleterious mutations such as transcriptional adaptation of compensatory genes, alternative mRNA splicing, and maternal deposition of wild-type transcripts, which are not observed in our system. The selective breeding approach and the resultant genetic circuitry change we uncovered advances and expands our current understanding of genetic and developmental resilience. Some deleterious gene mutations only affect a subset of genetically mutant animals. This widespread phenomenon, known as mutant incomplete penetrance, complicates discovery of causative gene mutations in both model organisms and human disease. This study utilized the zebrafish mef2ca transcription factor mutant that produces craniofacial skeleton defects with incomplete penetrance. Selectively breeding zebrafish families for low- or high-penetrance mutants for many generations created different zebrafish strains with consistently low or high penetrance. Comparing these strains allowed us to gain insight into the mechanisms that control penetrance. Specifically, genes under the control of mef2ca are initially similarly expressed between the two strains, but differences between strains emerge during development. We found that genetic manipulation of these downstream genes mimics the effects of our selective breeding. Thus, selective breeding for penetrance can change the genetic circuitry downstream of the mutated gene. We propose that small differences in gene circuitry between individuals is one mechanism underlying susceptibility or resilience to genetic mutations.
Collapse
Affiliation(s)
- Juliana Sucharov
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Kuval Ray
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Elliott P. Brooks
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - James T. Nichols
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
8
|
Hu X, Qin S, Huang X, Yuan Y, Tan Z, Gu Y, Cheng X, Wang D, Lian XF, He C, Su Z. Region-Restrict Astrocytes Exhibit Heterogeneous Susceptibility to Neuronal Reprogramming. Stem Cell Reports 2019; 12:290-304. [PMID: 30713039 PMCID: PMC6373495 DOI: 10.1016/j.stemcr.2018.12.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/26/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022] Open
Abstract
The adult CNS has poor ability to replace degenerated neurons following injury or disease. Recently, direct reprogramming of astrocytes into induced neurons has been proposed as an innovative strategy toward CNS repair. As a cell population that shows high diversity on physiological properties and functions depending on their spatiotemporal distribution, however, whether the astrocyte heterogeneity affect neuronal reprogramming is not clear. Here, we show that astrocytes derived from cortex, cerebellum, and spinal cord exhibit biological heterogeneity and possess distinct susceptibility to transcription factor-induced neuronal reprogramming. The heterogeneous expression level of NOTCH1 signaling in the different CNS regions-derived astrocytes is shown to be responsible for the neuronal reprogramming diversity. Taken together, our findings demonstrate that region-restricted astrocytes reveal different intrinsic limitation of the response to neuronal reprogramming. Region-restrict astrocytes (ACs) exhibit obvious heterogeneity Region-restrict ACs show distinct susceptibility to neuronal reprogramming AC heterogeneity does not affect the maturation of induced neurons Notch1 is involved in the neuronal reprogramming diversity of region-restrict ACs
Collapse
Affiliation(s)
- Xin Hu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China; Department of Neurological Surgery, Xixi Hospital of Hangzhou, Hangzhou 200233 China
| | - Shangyao Qin
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Xiao Huang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Yimin Yuan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Zijian Tan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Yakun Gu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Xueyan Cheng
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Dan Wang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China
| | - Xiao-Feng Lian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 310009, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China.
| | - Zhida Su
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
9
|
Jiao Y, Huang B, Chen Y, Hong G, Xu J, Hu C, Wang C. Integrated Analyses Reveal Overexpressed Notch1 Promoting Porcine Satellite Cells' Proliferation through Regulating the Cell Cycle. Int J Mol Sci 2018; 19:ijms19010271. [PMID: 29337929 PMCID: PMC5796217 DOI: 10.3390/ijms19010271] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 12/19/2022] Open
Abstract
Notch signaling as a conserved cell fate regulator is involved in the regulation of cell quiescence, proliferation, differentiation and postnatal tissue regeneration. However, how Notch signaling regulates porcine satellite cells (PSCs) has not been elucidated. We stably transfected Notch1 intracellular domain (N1ICD) into PSCs to analyze the gene expression profile and miRNA-seq. The analysis of the gene expression profile identified 295 differentially-expressed genes (DEGs) in proliferating-N1ICD PSCs (P-N1ICD) and nine DEGs on differentiating-N1ICD PSCs (D-N1ICD), compared with that in control groups (P-Control and D-Control, respectively). Analyzing the underlying function of DEGs showed that most of the upregulated DEGs enriched in P-N1ICD PSCs are related to the cell cycle. Forty-four and 12 known differentially-expressed miRNAs (DEMs) were identified in the P-N1ICD PSCs and D-N1ICD PSCs group, respectively. Furthermore, we constructed the gene-miRNA network of the DEGs and DEMs. In P-N1ICD PSCs, miR-125a, miR-125b, miR-10a-5p, ssc-miR-214, miR-423 and miR-149 are downregulated hub miRNAs, whose corresponding hub genes are marker of proliferation Ki-67 (MKI67) and nuclear receptor binding SET domain protein 2 (WHSC1). By contrast, miR-27a, miR-146a-5p and miR-221-3p are upregulated hub miRNAs, whose hub genes are RUNX1 translocation partner 1 (RUNX1T1) and fibroblast growth factor 2 (FGF2). All the hub miRNAs and genes are associated with cell proliferation. Quantitative RT-PCR results are consistent with the gene expression profile and miRNA-seq results. The results of our study provide valuable information for understanding the molecular mechanisms underlying Notch signaling in PSCs and skeletal muscle development.
Collapse
Affiliation(s)
- Yiren Jiao
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Bo Huang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Yu Chen
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Guangliang Hong
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Jian Xu
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| | - Chingyuan Hu
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, 1955 East-West Road, AgSci. 415J, Honolulu, HI 96822, USA.
| | - Chong Wang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
10
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 674] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
11
|
miR-142-3p Contributes to Early Cardiac Fate Decision of Embryonic Stem Cells. Stem Cells Int 2017; 2017:1769298. [PMID: 28656050 PMCID: PMC5474537 DOI: 10.1155/2017/1769298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/09/2017] [Indexed: 01/10/2023] Open
Abstract
MicroRNAs (miRNAs) play important roles in cell fate decisions. However, the miRNAs and their targets involved in the regulation of cardiac lineage specification are largely unexplored. Here, we report novel functions of miR-142-3p in the regulation of cardiomyocyte differentiation from mouse embryonic stem cells (mESCs). With a miRNA array screen, we identified a number of miRNAs significantly changed during mESC differentiation into the mesodermal and cardiac progenitor cells, and miR-142-3p was one among the markedly downregulated miRNAs. Ectopic expression and inhibition of miR-142-3p did not alter the characteristics of undifferentiated ESCs, whereas ectopic expression of miR-142-3p impaired cardiomyocyte formation. In addition, ectopic expression of miR-142-3p inhibited the expression of a cardiac mesodermal marker gene Mesp1 and downstream cardiac transcription factors Nkx2.5, Tbx5, and Mef2c but not the expression of three germ layer-specific genes. We further demonstrated that miR-142-3p targeted the 3'-untranslated region of Mef2c. These results reveal miR-142-3p as an important regulator of early cardiomyocyte differentiation. Our findings provide new knowledge for further understanding of roles and mechanisms of miRNAs as critical regulators of cardiomyocyte differentiation.
Collapse
|
12
|
Desjardins CA, Naya FJ. Antagonistic regulation of cell-cycle and differentiation gene programs in neonatal cardiomyocytes by homologous MEF2 transcription factors. J Biol Chem 2017; 292:10613-10629. [PMID: 28473466 DOI: 10.1074/jbc.m117.776153] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 05/03/2017] [Indexed: 12/30/2022] Open
Abstract
Cardiomyocytes acquire their primary specialized function (contraction) before exiting the cell cycle. In this regard, proliferation and differentiation must be precisely coordinated for proper cardiac morphogenesis. Here, we have investigated the complex transcriptional mechanisms employed by cardiomyocytes to coordinate antagonistic cell-cycle and differentiation gene programs through the molecular dissection of the core cardiac transcription factor, MEF2. Knockdown of individual MEF2 proteins, MEF2A, -C, and -D, in primary neonatal cardiomyocytes resulted in radically distinct and opposite effects on cellular homeostasis and gene regulation. MEF2A and MEF2D were absolutely required for cardiomyocyte survival, whereas MEF2C, despite its major role in cardiac morphogenesis and direct reprogramming, was dispensable for this process. Inhibition of MEF2A or -D also resulted in the activation of cell-cycle genes and down-regulation of markers of terminal differentiation. In striking contrast, the regulation of cell-cycle and differentiation gene programs by MEF2C was antagonistic to that of MEF2A and -D. Computational analysis of regulatory regions from MEF2 isoform-dependent gene sets identified the Notch and Hedgehog signaling pathways as key determinants in coordinating MEF2 isoform-specific control of antagonistic gene programs. These results reveal that mammalian MEF2 family members have distinct transcriptional functions in cardiomyocytes and suggest that these differences are critical for proper development and maturation of the heart. Analysis of MEF2 isoform-specific function in neonatal cardiomyocytes has yielded insight into an unexpected transcriptional regulatory mechanism by which these specialized cells utilize homologous members of a core cardiac transcription factor to coordinate cell-cycle and differentiation gene programs.
Collapse
Affiliation(s)
- Cody A Desjardins
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| | - Francisco J Naya
- From the Department of Biology, Program in Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215
| |
Collapse
|
13
|
Shan T, Xu Z, Wu W, Liu J, Wang Y. Roles of Notch1 Signaling in Regulating Satellite Cell Fates Choices and Postnatal Skeletal Myogenesis. J Cell Physiol 2017; 232:2964-2967. [DOI: 10.1002/jcp.25730] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 12/07/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Tizhong Shan
- College of Animal Sciences; Zhejiang University; Hangzhou Zhejiang P. R. China
- The Key Laboratory of Molecular Animal Nutrition; Ministry of Education; Hangzhou Zhejiang P. R. China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang P. R. China
| | - Ziye Xu
- College of Animal Sciences; Zhejiang University; Hangzhou Zhejiang P. R. China
- The Key Laboratory of Molecular Animal Nutrition; Ministry of Education; Hangzhou Zhejiang P. R. China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang P. R. China
| | - Weiche Wu
- College of Animal Sciences; Zhejiang University; Hangzhou Zhejiang P. R. China
- The Key Laboratory of Molecular Animal Nutrition; Ministry of Education; Hangzhou Zhejiang P. R. China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang P. R. China
| | - Jiaqi Liu
- College of Animal Sciences; Zhejiang University; Hangzhou Zhejiang P. R. China
- The Key Laboratory of Molecular Animal Nutrition; Ministry of Education; Hangzhou Zhejiang P. R. China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang P. R. China
| | - Yizhen Wang
- College of Animal Sciences; Zhejiang University; Hangzhou Zhejiang P. R. China
- The Key Laboratory of Molecular Animal Nutrition; Ministry of Education; Hangzhou Zhejiang P. R. China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition; Hangzhou Zhejiang P. R. China
| |
Collapse
|
14
|
Abad M, Hashimoto H, Zhou H, Morales MG, Chen B, Bassel-Duby R, Olson EN. Notch Inhibition Enhances Cardiac Reprogramming by Increasing MEF2C Transcriptional Activity. Stem Cell Reports 2017; 8:548-560. [PMID: 28262548 PMCID: PMC5355682 DOI: 10.1016/j.stemcr.2017.01.025] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 01/14/2023] Open
Abstract
Conversion of fibroblasts into functional cardiomyocytes represents a potential means of restoring cardiac function after myocardial infarction, but so far this process remains inefficient and little is known about its molecular mechanisms. Here we show that DAPT, a classical Notch inhibitor, enhances the conversion of mouse fibroblasts into induced cardiac-like myocytes by the transcription factors GATA4, HAND2, MEF2C, and TBX5. DAPT cooperates with AKT kinase to further augment this process, resulting in up to 70% conversion efficiency. Moreover, DAPT promotes the acquisition of specific cardiomyocyte features, substantially increasing calcium flux, sarcomere structure, and the number of spontaneously beating cells. Transcriptome analysis shows that DAPT induces genetic programs related to muscle development, differentiation, and excitation-contraction coupling. Mechanistically, DAPT increases binding of the transcription factor MEF2C to the promoter regions of cardiac structural genes. These findings provide mechanistic insights into the reprogramming process and may have important implications for cardiac regeneration therapies. Notch activation is a barrier for GHMT-induced cardiac cell reprogramming Notch blockade by DAPT improves GHMT-induced cardiac reprogramming DAPT increases sarcomere organization, calcium flux, and beating in GHMT reprogramming DAPT enhances transcriptional activity of MEF2C in GHMT reprogramming
Collapse
Affiliation(s)
- Maria Abad
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Cell Plasticity and Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), c/Natzaret, 115-117, Barcelona 08035, Spain.
| | - Hisayuki Hashimoto
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Huanyu Zhou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Maria Gabriela Morales
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Beibei Chen
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
15
|
Abstract
Diseases of muscle that are caused by pathological interactions between muscle and the immune system are devastating, but rare. However, muscle injuries that involve trauma and regeneration are fairly common, and inflammation is a clear feature of the regenerative process. Investigations of the inflammatory response to muscle injury have now revealed that the apparently nonspecific inflammatory response to trauma is actually a complex and coordinated interaction between muscle and the immune system that determines the success or failure of tissue regeneration.
Collapse
Affiliation(s)
- James G Tidball
- Molecular, Cellular and Integrative Physiology Program, University of California, Los Angeles, California 90095-1606, USA
| |
Collapse
|
16
|
Kim YJ, Tamadon A, Park HT, Kim H, Ku SY. The role of sex steroid hormones in the pathophysiology and treatment of sarcopenia. Osteoporos Sarcopenia 2016; 2:140-155. [PMID: 30775480 PMCID: PMC6372754 DOI: 10.1016/j.afos.2016.06.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/09/2016] [Accepted: 06/17/2016] [Indexed: 12/18/2022] Open
Abstract
Sex steroids influence the maintenance and growth of muscles. Decline in androgens, estrogens and progesterone by aging leads to the loss of muscular function and mass, sarcopenia. These steroid hormones can interact with different signaling pathways through their receptors. To date, sex steroid hormone receptors and their exact roles are not completely defined in skeletal and smooth muscles. Although numerous studies focused on the effects of sex steroid hormones on different types of cells, still many unexplained molecular mechanisms in both skeletal and smooth muscle cells remain to be investigated. In this paper, many different molecular mechanisms that are activated or inhibited by sex steroids and those that influence the growth, proliferation, and differentiation of skeletal and smooth muscle cells are reviewed. Also, the similarities of cellular and molecular pathways of androgens, estrogens and progesterone in both skeletal and smooth muscle cells are highlighted. The reviewed signaling pathways and participating molecules can be targeted in the future development of novel therapeutics.
Collapse
Affiliation(s)
- Yong Jin Kim
- Department of Obstetrics and Gynecology, Korea University Guro Hospital, South Korea
| | - Amin Tamadon
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Hyun Tae Park
- Department of Obstetrics and Gynecology, Korea University Anam Hospital, Korea University College of Medicine, South Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
17
|
Villasante A, Powell MS, Murdoch GK, Overturf K, Cain K, Wacyk J, Hardy RW. Effect of anthocyanidins on myogenic differentiation in induced and non-induced primary myoblasts from rainbow trout ( Oncorhynchus mykiss ). Comp Biochem Physiol B Biochem Mol Biol 2016; 196-197:102-108. [DOI: 10.1016/j.cbpb.2016.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/03/2016] [Accepted: 03/07/2016] [Indexed: 10/22/2022]
|
18
|
|
19
|
Shan T, Zhang P, Xiong Y, Wang Y, Kuang S. Lkb1 deletion upregulates Pax7 expression through activating Notch signaling pathway in myoblasts. Int J Biochem Cell Biol 2016; 76:31-8. [PMID: 27131604 DOI: 10.1016/j.biocel.2016.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 03/19/2016] [Accepted: 04/26/2016] [Indexed: 11/28/2022]
Abstract
Satellite cells play crucial roles in mediating the growth, maintenance, and repair of postnatal skeletal muscle. Activated satellite cells (myoblasts) can divide symmetrically or asymmetrically to generate progenies that self-renewal, proliferate or differentiate. Pax7 is a defining marker of quiescent and activated satellite cells, but not differentiated myoblast. We demonstrate here that deletion of Lkb1 upregulates Pax7 expression in myoblasts and inhibits asymmetric divisions that generate differentiating progenies. Furthermore, we find that Lkb1 activates the Notch signaling pathway, which subsequently increases Pax7 expression and promotes self-renewal and proliferation while inhibiting differentiation. Mechanistic studies reveal that Lkb1 regulates Notch activation through AMPK-mTOR pathway in myoblasts. Together, these results establish a key role of Lkb1 in regulating myoblast division and cell fates choices.
Collapse
Affiliation(s)
- Tizhong Shan
- Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA.
| | - Pengpeng Zhang
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| | - Yan Xiong
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA
| | - Yizhen Wang
- Department of Animal Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shihuan Kuang
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA.
| |
Collapse
|
20
|
Jin W, Peng J, Jiang S. The epigenetic regulation of embryonic myogenesis and adult muscle regeneration by histone methylation modification. Biochem Biophys Rep 2016; 6:209-219. [PMID: 28955879 PMCID: PMC5600456 DOI: 10.1016/j.bbrep.2016.04.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle formation in vertebrates is derived from the paraxial mesoderm, which develops into myogenic precursor cells and finally differentiates into mature myofibers. This myogenic program involves temporal-spatial molecular events performed by transcription regulators (such as members of the Pax, MRFs and Six families) and signaling pathways (such as Wnts, BMP and Shh signaling). Epigenetic regulation, including histone post-translational modifications is crucial for controlling gene expression through recruitment of various chromatin-modifying enzymes that alter chromatin dynamics during myogenesis. The chromatin modifying enzymes are also recruited at regions of muscle gene regulation, coordinating transcription regulators to influence gene expression. In particular, the reversible methylation status of histone N-terminal tails provides the important regulatory mechanisms in either activation or repression of muscle genes. In this report, we review the recent literatures to deduce mechanisms underlying the epigenetic regulation of gene expression with a focus on histone methylation modification during embryo myogenesis and adult muscle regeneration. Recent results from different histone methylation/demethylation modifications have increased our understanding about the highly intricate layers of epigenetic regulations involved in myogenesis and cross-talk of histone enzymes with the muscle-specific transcriptional machinery. Myogenesis is influenced by regulation of transcription factors, signal pathways and post-transcriptional modifications. Histone methylation modifications as “on/off” switches regulated myogenic lineage commitment and differentiation. The myogenic regulatory factors and histone methylation modifications established dynamic regulatory mechanism.
Collapse
Key Words
- BMP4, bone morphogenic protein 4
- ChIP, chromatin immunoprecipitation
- Epigenetic
- H3K27, methylation of histone H3 lysine 27
- H3K4, methylation of histone H3 lysine 4
- H3K9, methylation of histone H3 lysine 9
- Histone methylation/demethylation modification
- KDMs, lysine demethyltransferases
- LSD1, lysine specific demethyltransferase 1
- MEF2, myocyte enhancer factor 2
- MRFs, myogenic regulatory factors
- Muscle differentiation
- Muscle progenitor cells
- Muscle regeneration
- Myogenesis
- PRC2, polycomb repressive complex 2
- SCs, satellite cells
- Shh, sonic hedgehog
- TSS, transcription start sites
- UTX, ubiquitously transcribed tetratricopeptide repeat, X chromosome
- bHLH, basic helix-loop-helix
- p38 MAPK, p38 mitogen-activated protein kinase
Collapse
Affiliation(s)
- Wei Jin
- Key Laboratory of Pig Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, PR China
| | - Siwen Jiang
- Key Laboratory of Pig Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, PR China.,Key Projects in the Cooperative Innovation Center for Sustainable Pig Production of Wuhan, PR China
| |
Collapse
|
21
|
Cao XK, Zhan ZY, Huang YZ, Lan XY, Lei CZ, Qi XL, Chen H. Variants and haplotypes within MEF2C gene influence stature of chinese native cattle including body dimensions and weight. Livest Sci 2016. [DOI: 10.1016/j.livsci.2016.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
22
|
Muscle-specific microRNAs in skeletal muscle development. Dev Biol 2016; 410:1-13. [DOI: 10.1016/j.ydbio.2015.12.013] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 01/19/2023]
|
23
|
Abstract
The developmental mechanisms that control head muscle formation are distinct from those that operate in the trunk. Head and neck muscles derive from various mesoderm populations in the embryo and are regulated by distinct transcription factors and signaling molecules. Throughout the last decade, developmental, and lineage studies in vertebrates and invertebrates have revealed the peculiar nature of the pharyngeal mesoderm that forms certain head muscles and parts of the heart. Studies in chordates, the ancestors of vertebrates, revealed an evolutionarily conserved cardiopharyngeal field that progressively facilitates the development of both heart and craniofacial structures during vertebrate evolution. This ancient regulatory circuitry preceded and facilitated the emergence of myogenic cell types and hierarchies that exist in vertebrates. This chapter summarizes studies related to the origins, signaling circuits, genetics, and evolution of the head musculature, highlighting its heterogeneous characteristics in all these aspects, with a special focus on the FGF-ERK pathway. Additionally, we address the processes of head muscle regeneration, and the development of stem cell-based therapies for treatment of muscle disorders.
Collapse
Affiliation(s)
- Inbal Michailovici
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Eigler
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
24
|
George RM, Hahn KL, Rawls A, Viger RS, Wilson-Rawls J. Notch signaling represses GATA4-induced expression of genes involved in steroid biosynthesis. Reproduction 2015; 150:383-94. [PMID: 26183893 DOI: 10.1530/rep-15-0226] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/16/2015] [Indexed: 12/18/2022]
Abstract
Notch2 and Notch3 and genes of the Notch signaling network are dynamically expressed in developing follicles, where they are essential for granulosa cell proliferation and meiotic maturation. Notch receptors, ligands, and downstream effector genes are also expressed in testicular Leydig cells, predicting a potential role in regulating steroidogenesis. In this study, we sought to determine if Notch signaling in small follicles regulates the proliferation response of granulosa cells to FSH and represses the up-regulation steroidogenic gene expression that occurs in response to FSH as the follicle grows. Inhibition of Notch signaling in small preantral follicles led to the up-regulation of the expression of genes in the steroid biosynthetic pathway. Similarly, progesterone secretion by MA-10 Leydig cells was significantly inhibited by constitutively active Notch. Together, these data indicated that Notch signaling inhibits steroidogenesis. GATA4 has been shown to be a positive regulator of steroidogenic genes, including STAR protein, P450 aromatase, and 3B-hydroxysteroid dehydrogenase. We observed that Notch downstream effectors HEY1, HEY2, and HEYL are able to differentially regulate these GATA4-dependent promoters. These data are supported by the presence of HEY/HES binding sites in these promoters. These studies indicate that Notch signaling has a role in the complex regulation of the steroidogenic pathway.
Collapse
Affiliation(s)
- Rajani M George
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Katherine L Hahn
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Alan Rawls
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Robert S Viger
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4 School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| | - Jeanne Wilson-Rawls
- School of Life SciencesArizona State University, PO Box 874501, Tempe, Arizona 85827-45012, USAReproductionMother and Child Health, Centre de Recherche du CHU de Québec and Centre de Recherche en Biologie de la Reproduction (CRBR), Quebec City, Quebec, CanadaDepartment of ObstetricsGynecology, and Reproduction, Laval University, Quebec City, Quebec, Canada G1K 7P4
| |
Collapse
|
25
|
Castells-Sala C, Martínez-Ramos C, Vallés-Lluch A, Monleón Pradas M, Semino C. in vitro development of bioimplants made up of elastomeric scaffolds with peptide gel filling seeded with human subcutaneous adipose tissue-derived progenitor cells. J Biomed Mater Res A 2015; 103:3419-30. [PMID: 25903327 DOI: 10.1002/jbm.a.35482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 03/31/2015] [Accepted: 04/13/2015] [Indexed: 12/17/2022]
Abstract
Myocardial tissue lacks the ability to regenerate itself significantly following a myocardial infarction. Thus, new strategies that could compensate this lack are of high interest. Cardiac tissue engineering (CTE) strategies are a relatively new approach that aims to compensate the tissue loss using combination of biomaterials, cells and bioactive molecules. The goal of the present study was to evaluate cell survival and growth, seeding capacity and cellular phenotype maintenance of subcutaneous adipose tissue-derived progenitor cells in a new synthetic biomaterial scaffold platform. Specifically, here we tested the effect of the RAD16-I peptide gel in microporous poly(ethyl acrylate) polymers using two-dimensional PEA films as controls. Results showed optimal cell adhesion efficiency and growth in the polymers coated with the self-assembling peptide RAD16-I. Importantly, subATDPCs seeded into microporous PEA scaffolds coated with RAD16-I maintained its phenotype and were able to migrate outwards the bioactive patch, hopefully toward the infarcted area once implanted. These data suggest that this bioimplant (scaffold/RAD16-I/cells) can be suitable for further in vivo implantation with the aim to improve the function of affected tissue after myocardial infarction.
Collapse
Affiliation(s)
- Cristina Castells-Sala
- Tissue Engineering Laboratory, Bioengineering Department, Institut Químic De Sarrià, Universitat Ramon Llull, Barcelona, Spain
| | - Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica De Valencia, Cno. De Vera S/N, Valencia, 46022, Spain
| | - Ana Vallés-Lluch
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica De Valencia, Cno. De Vera S/N, Valencia, 46022, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica De Valencia, Cno. De Vera S/N, Valencia, 46022, Spain
| | - Carlos Semino
- Tissue Engineering Laboratory, Bioengineering Department, Institut Químic De Sarrià, Universitat Ramon Llull, Barcelona, Spain
| |
Collapse
|
26
|
Abstract
Embryonal rhabdomyosarcoma is one of the major defined histologic variants of rhabdomyosarcoma that is mainly reported in children. The histologic appearance of this neoplastic entity recapitulates normal myogenesis. The tumor cells variably exhibit the different cellular phases of myogenesis ranging from undifferentiated mesenchymal cells to elongated myoblasts, multinucleated myotubes and differentiated muscle fibers. The carefully orchestrated embryonic signaling pathways that are involved in myogenesis, conceivably also result in the genesis of rhabdomyosarcoma; albeit as a corollary to an imbalance. We have attempted to review the pathogenesis of embryonal rhabdomyosarcoma in an endeavor to understand better, how closely it is linked to normal myogenesis in terms of its molecular dynamics and histologic presentation.
Collapse
|
27
|
Zhang M, Zhu B, Davie J. Alternative splicing of MEF2C pre-mRNA controls its activity in normal myogenesis and promotes tumorigenicity in rhabdomyosarcoma cells. J Biol Chem 2014; 290:310-24. [PMID: 25404735 DOI: 10.1074/jbc.m114.606277] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma in children. Many cellular disruptions contribute to the progression of this pediatric cancer, including aberrant alternative splicing. The MEF2 family of transcription factors regulates many developmental programs, including myogenesis. MEF2 gene transcripts are subject to alternate splicing to generate protein isoforms with divergent functions. We found that MEF2Cα1 was the ubiquitously expressed isoform that exhibited no myogenic activity and that MEF2Cα2, the muscle-specific MEF2C isoform, was required for efficient differentiation. We showed that exon α in MEF2C was aberrantly alternatively spliced in RMS cells, with the ratio of α2/α1 highly down-regulated in RMS cells compared with normal myoblasts. Compared with MEF2Cα2, MEF2Cα1 interacted more strongly with and recruited HDAC5 to myogenic gene promoters to repress muscle-specific genes. Overexpression of the MEF2Cα2 isoform in RMS cells increased myogenic activity and promoted differentiation in RMS cells. We also identified a serine protein kinase, SRPK3, that was down-regulated in RMS cells and found that expression of SRPK3 promoted the splicing of the MEF2Cα2 isoform and induced differentiation. Restoration of either MEF2Cα2 or SPRK3 inhibited both proliferation and anchorage-independent growth of RMS cells. Together, our findings indicate that the alternative splicing of MEF2C plays an important role in normal myogenesis and RMS development. An improved understanding of alternative splicing events in RMS cells will potentially reveal novel therapeutic targets for RMS treatment.
Collapse
Affiliation(s)
- Meiling Zhang
- From the Department of Biochemistry and Molecular Biology and Simmons Cancer Institute, Southern Illinois University, School of Medicine, Carbondale, Illinois 62901
| | - Bo Zhu
- From the Department of Biochemistry and Molecular Biology and Simmons Cancer Institute, Southern Illinois University, School of Medicine, Carbondale, Illinois 62901
| | - Judith Davie
- From the Department of Biochemistry and Molecular Biology and Simmons Cancer Institute, Southern Illinois University, School of Medicine, Carbondale, Illinois 62901
| |
Collapse
|
28
|
Hutchins ED, Markov GJ, Eckalbar WL, George RM, King JM, Tokuyama MA, Geiger LA, Emmert N, Ammar MJ, Allen AN, Siniard AL, Corneveaux JJ, Fisher RE, Wade J, DeNardo DF, Rawls JA, Huentelman MJ, Wilson-Rawls J, Kusumi K. Transcriptomic analysis of tail regeneration in the lizard Anolis carolinensis reveals activation of conserved vertebrate developmental and repair mechanisms. PLoS One 2014; 9:e105004. [PMID: 25140675 PMCID: PMC4139331 DOI: 10.1371/journal.pone.0105004] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 07/17/2014] [Indexed: 01/09/2023] Open
Abstract
Lizards, which are amniote vertebrates like humans, are able to lose and regenerate a functional tail. Understanding the molecular basis of this process would advance regenerative approaches in amniotes, including humans. We have carried out the first transcriptomic analysis of tail regeneration in a lizard, the green anole Anolis carolinensis, which revealed 326 differentially expressed genes activating multiple developmental and repair mechanisms. Specifically, genes involved in wound response, hormonal regulation, musculoskeletal development, and the Wnt and MAPK/FGF pathways were differentially expressed along the regenerating tail axis. Furthermore, we identified 2 microRNA precursor families, 22 unclassified non-coding RNAs, and 3 novel protein-coding genes significantly enriched in the regenerating tail. However, high levels of progenitor/stem cell markers were not observed in any region of the regenerating tail. Furthermore, we observed multiple tissue-type specific clusters of proliferating cells along the regenerating tail, not localized to the tail tip. These findings predict a different mechanism of regeneration in the lizard than the blastema model described in the salamander and the zebrafish, which are anamniote vertebrates. Thus, lizard tail regrowth involves the activation of conserved developmental and wound response pathways, which are potential targets for regenerative medical therapies.
Collapse
Affiliation(s)
- Elizabeth D. Hutchins
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Glenn J. Markov
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Walter L. Eckalbar
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Rajani M. George
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Jesse M. King
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Minami A. Tokuyama
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Lauren A. Geiger
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Nataliya Emmert
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Michael J. Ammar
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - April N. Allen
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Ashley L. Siniard
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jason J. Corneveaux
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Rebecca E. Fisher
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
| | - Juli Wade
- Departments of Psychology and Zoology, Program in Neuroscience, Michigan State University, East Lansing, Michigan, United States of America
| | - Dale F. DeNardo
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - J. Alan Rawls
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Matthew J. Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Kenro Kusumi
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, Arizona, United States of America
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona, United States of America
| |
Collapse
|
29
|
Fujiwara T, Kunisada T, Takeda K, Uotani K, Yoshida A, Ochiya T, Ozaki T. MicroRNAs in soft tissue sarcomas: overview of the accumulating evidence and importance as novel biomarkers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:592868. [PMID: 25165708 PMCID: PMC4139009 DOI: 10.1155/2014/592868] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/09/2014] [Indexed: 12/11/2022]
Abstract
Sarcomas are distinctly heterogeneous tumors and a variety of subtypes have been described. Although several diagnostic explorations in the past three decades, such as identification of chromosomal translocation, have greatly improved the diagnosis of soft tissue sarcomas, the unsolved issues, including the limited useful biomarkers, remain. Emerging reports on miRNAs in soft tissue sarcomas have provided clues to solving these problems. Evidence of circulating miRNAs in patients with soft tissue sarcomas and healthy individuals has been accumulated and is accelerating their potential to develop into clinical applications. Moreover, miRNAs that function as novel prognostic factors have been identified, thereby facilitating their use in miRNA-targeted therapy. In this review, we provide an overview of the current knowledge on miRNA deregulation in soft tissue sarcomas, and discuss their potential as novel biomarkers and therapeutics.
Collapse
Affiliation(s)
- Tomohiro Fujiwara
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 7008558, Japan
| | - Toshiyuki Kunisada
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
- Department of Medical Materials for Musculoskeletal Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Ken Takeda
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
- Department of Intelligent Orthopaedic System, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Koji Uotani
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Aki Yoshida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 1040045, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
| |
Collapse
|
30
|
Sturtzel C, Testori J, Schweighofer B, Bilban M, Hofer E. The transcription factor MEF2C negatively controls angiogenic sprouting of endothelial cells depending on oxygen. PLoS One 2014; 9:e101521. [PMID: 24988463 PMCID: PMC4079651 DOI: 10.1371/journal.pone.0101521] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/09/2014] [Indexed: 11/18/2022] Open
Abstract
The MADS box transcription factor MEF2C has been detected by us to be upregulated by the angiogenic factors VEGF-A and bFGF in endothelial cells. We have here investigated its potential role for angiogenesis. MEF2C was surprisingly found to strongly inhibit angiogenic sprouting, whereas a dominant negative mutant rather induced sprouting. The factor mainly affected migratory processes of endothelial cells, but not proliferation. In gene profiling experiments we delineated the alpha-2-macroglobulin gene to be highly upregulated by MEF2C. Further data confirmed that MEF2C in endothelial cells indeed induces alpha-2-macroglobulin mRNA as well as the secretion of alpha-2-macroglobulin and that conditioned supernatants of cells overexpressing MEF2C inhibit sprouting. Alpha-2-macroglobulin mediates, at least to a large extent, the inhibitory effects of MEF2C as is shown by knockdown of alpha-2-macroglobulin mRNA by lentiviral shRNA expression which reduces the inhibitory effect. However, under hypoxic conditions the VEGF-A/bFGF-mediated upregulation of MEF2C is reduced and the production of alpha-2-macroglobulin largely abolished. Taken together, this suggests that the MEF2C/alpha-2-macroglobulin axis functions in endothelial cells as a negative feed-back mechanism that adapts sprouting activity to the oxygen concentration thus diminishing inappropriate and excess angiogenesis.
Collapse
Affiliation(s)
- Caterina Sturtzel
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julia Testori
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Schweighofer
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Erhard Hofer
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
31
|
Khanna N, Ge Y, Chen J. MicroRNA-146b promotes myogenic differentiation and modulates multiple gene targets in muscle cells. PLoS One 2014; 9:e100657. [PMID: 24956113 PMCID: PMC4067360 DOI: 10.1371/journal.pone.0100657] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/26/2014] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs are established as crucial modulators of skeletal myogenesis, but our knowledge about their identity and targets remains limited. In this study, we have identified microRNA-146b (miR-146b) as a novel regulator of skeletal myoblast differentiation. Following up on a previous microRNA profiling study, we establish that the expression of miR-146b is up-regulated during myoblast differentiation in vitro and muscle regeneration in vivo. Inhibition of miR-146b led to reduced myoblast differentiation, whereas overexpression of miR-146b enhanced differentiation. Computational prediction combined with gene expression information has revealed candidates for miR-146b targets in muscles. Among them, the expression of Smad4, Notch1, and Hmga2 are significantly suppressed by miR-146b overexpression in myocytes. In addition, expression levels of Smad4, Notch1 and Hmga2 are decreased during myoblast differentiation and muscle regeneration, inversely correlating to the levels of miR-146b. Importantly, inhibition of endogenous miR-146b prevents the down-regulation of Smad4, Notch1 and Hmga2 during differentiation. Furthermore, miR-146b directly targets the microRNA response elements (MREs) in the 3'UTR of those genes as assessed by reporter assays. Reporters with the seed regions of MREs mutated are insensitive to miR-146b, further confirming the specificity of targeting. In conclusion, miR-146b is a positive regulator of myogenic differentiation, possibly acting through multiple targets.
Collapse
Affiliation(s)
- Nidhi Khanna
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yejing Ge
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jie Chen
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
32
|
Wen F, Wong HK, Tay CY, Yu H, Li H, Yu T, Tijore A, Boey FYC, Venkatraman SS, Tan LP. Induction of myogenic differentiation of human mesenchymal stem cells cultured on Notch agonist (Jagged-1) modified biodegradable scaffold surface. ACS APPLIED MATERIALS & INTERFACES 2014; 6:1652-61. [PMID: 24405311 DOI: 10.1021/am4045635] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Engineered scaffold surface provides stem cells with vital cues that could determine the eventual fate of stem cells. In this work, biodegradable poly(L-lactide-co-ε-caprolactone) (PLCL) scaffold conjugated with Notch agonist-Jagged-1(JAG) peptide (2.1 kDa) was prepared to initiate myogenic differentiation of human mesenchymal stem cells (hMSCs). The scaffold surface was activated with oxygen plasma and acrylic acid was engrafted via UV polymerization to form a surface bearing carboxylic groups. JAG peptide was subsequently immobilized onto the carboxylated scaffold surface. Surface chemistry and topography were examined using attenuated total reflection Fourier transform infrared, X-ray photoelectron spectroscopy, and atomic force microscopy. Quantitative real time polymerase chain reaction analysis revealed activation of the Notch pathway; furthermore, several specific markers associated with myogenic but not osteogenic differentiation were shown to be up-regulated in hMSCs cultured on the engineered surface. The pro-myocardial effect of surface bound JAG peptide was further affirmed via immunodetection of the distinct myocardial marker, cardiac troponin T. Collectively, our results suggest that PLCL conjugated JAG peptide is a viable strategy to enhance the functional potential of scaffolds to be used as a bioengineered cardiac patch in myocardial infarction repair.
Collapse
Affiliation(s)
- Feng Wen
- Division of Materials Technology, School of Materials Science and Engineering, Nanyang Technological University , 50 Nanyang Avenue, 639798 Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Diao Y, Guo X, Jiang L, Wang G, Zhang C, Wan J, Jin Y, Wu Z. miR-203, a tumor suppressor frequently down-regulated by promoter hypermethylation in rhabdomyosarcoma. J Biol Chem 2013; 289:529-39. [PMID: 24247238 DOI: 10.1074/jbc.m113.494716] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma found in children and young adults. It is characterized by the expression of a number of skeletal muscle-specific proteins, including MyoD and muscle α-actin. However, unlike normal myoblasts, RMS cells differentiate poorly both in vivo and in culture. As microRNAs are known to regulate tumorigenesis, intensive efforts have been made to identify microRNAs that are involved in RMS development. In this work, we found that miR-203 was frequently down-regulated by promoter hypermethylation in both RMS cell lines and RMS biopsies and could be reactivated by DNA-demethylating agents. Re-expression of miR-203 in RMS cells inhibited their migration and proliferation and promoted terminal myogenic differentiation. Mechanistically, miR-203 exerts its tumor-suppressive effect by directly targeting p63 and leukemia inhibitory factor receptor in RMS cells, which promotes myogenic differentiation by inhibiting the Notch and the JAK1/STAT1/STAT3 pathways, respectively. Our work reveals that miR-203 functions as a tumor suppressor in RMS development.
Collapse
Affiliation(s)
- Yarui Diao
- From the Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, 518036 Shenzhen
| | | | | | | | | | | | | | | |
Collapse
|
34
|
MacGrogan D, Luxán G, de la Pompa JL. Genetic and functional genomics approaches targeting the Notch pathway in cardiac development and congenital heart disease. Brief Funct Genomics 2013; 13:15-27. [PMID: 24106100 DOI: 10.1093/bfgp/elt036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The Notch signalling pathway plays crucial roles in cardiac development and postnatal cardiac homoeostasis. Gain- and loss-of-function approaches indicate that Notch promotes or inhibits cardiogenesis in a stage-dependent manner. However, the molecular mechanisms are poorly defined because many downstream effectors remain to be identified. Genome-scale analyses are shedding light on the genes that are regulated by Notch signalling and the mechanisms underlying this regulation. We review the functional data that implicates Notch in cardiac morphogenetic processes and expression profiling studies that enlighten the regulatory networks behind them. A recurring theme is that Notch cross-talks reiteratively with other key signalling pathways including Wnt and Bmp to coordinate cell and tissue interactions during cardiogenesis.
Collapse
Affiliation(s)
- Donal MacGrogan
- Program of Cardiovascular Developmental Biology, Department of Cardiovascular Development and Repair, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain. Tel.: +34-620-936633; Fax: +34-91-4531304;
| | | | | |
Collapse
|
35
|
Abstract
Notch signaling has been shown over the past few decades to play fundamental roles in a plethora of developmental processes in an evolutionarily conserved fashion. Notch-mediated cell-to-cell signaling is involved in many aspects of embryonic development and control of tissue homeostasis in a variety of adult tissues, and regulates stem cell maintenance, cell differentiation and cellular homeostasis. The focus of this Review is the role of Notch signaling in stem cells, comparing insights from flies, fish and mice to highlight similarities, as well as differences, between species, tissues and stem cell compartments.
Collapse
Affiliation(s)
- Ute Koch
- Ecole Polytechnique Fédérale de Lausanne (EPFL), School of Life Science, SwissInstitute for Experimental Cancer Research (ISREC), Station 19, 1015 Lausanne, Switzerland.
| | | | | |
Collapse
|
36
|
Cappellari O, Benedetti S, Innocenzi A, Tedesco FS, Moreno-Fortuny A, Ugarte G, Lampugnani MG, Messina G, Cossu G. Dll4 and PDGF-BB convert committed skeletal myoblasts to pericytes without erasing their myogenic memory. Dev Cell 2013; 24:586-99. [PMID: 23477786 DOI: 10.1016/j.devcel.2013.01.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 12/21/2012] [Accepted: 01/28/2013] [Indexed: 11/25/2022]
Abstract
Pericytes are endothelial-associated cells that contribute to vessel wall. Here, we report that pericytes may derive from direct conversion of committed skeletal myoblasts. When exposed to Dll4 and PDGF-BB, but not Dll1, skeletal myoblasts downregulate myogenic genes, except Myf5, and upregulate pericyte markers, whereas inhibition of Notch signaling restores myogenesis. Moreover, when cocultured with endothelial cells, skeletal myoblasts, previously treated with Dll4 and PDGF-BB, adopt a perithelial position stabilizing newly formed vessel-like networks in vitro and in vivo. In a transgenic mouse model in which cells expressing MyoD activate Notch, skeletal myogenesis is abolished and pericyte genes are activated. Even if overexpressed, Myf5 does not trigger myogenesis because Notch induces Id3, partially sequestering Myf5 and inhibiting MEF2 expression. Myf5-expressing cells adopt a perithelial position, as occasionally also observed in wild-type (WT) embryos. These data indicate that endothelium, via Dll4 and PDGF-BB, induces a fate switch in adjacent skeletal myoblasts.
Collapse
Affiliation(s)
- Ornella Cappellari
- Department of Cell and Developmental Biology and Centre for Stem Cells and Regenerative Medicine, University College London, WC1E 6DE London, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Myocyte enhancer factor 2C in hematopoiesis and leukemia. Oncogene 2013; 33:403-10. [PMID: 23435431 DOI: 10.1038/onc.2013.56] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
Abstract
MEF2C is a selectively expressed transcription factor involved in different transcriptional complexes. Originally identified as an essential regulator of muscle development, ectopic expression of MEF2C as a result of chromosomal rearrangements is now linked to leukemia. Specifically, high MEF2C expression has been linked to mixed lineage leukemia-rearranged acute myeloid leukemia as well as to the immature subgroup of T-cell acute lymphoblastic leukemia. This review focuses on the role of MEF2C in the hematopoietic system and on aberrant MEF2C expression in human leukemia.
Collapse
|
38
|
Debnath I, Roundy KM, Pioli PD, Weis JJ, Weis JH. Bone marrow-induced Mef2c deficiency delays B-cell development and alters the expression of key B-cell regulatory proteins. Int Immunol 2012; 25:99-115. [PMID: 23087187 DOI: 10.1093/intimm/dxs088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The Mef2 family transcriptional regulator Mef2c (myocyte enhancer factor 2c) is highly expressed in maturing bone marrow and peripheral mature B-cells. To evaluate the role of this transcription factor in B-cell development, we generated a B-cell-specific conditional deletion of Mef2c using the Mb-1-Cre transgene that is expressed during the early stages of immunoglobulin rearrangement. Young mice possessing this defect demonstrated a significant impairment in B-cell numbers in bone marrow and spleen. This phenotype was evident in all B-cell subsets; however, as the animals mature, the deficit in the peripheral mature B-cell compartments was overcome. The absence of Mef2c in mature B-cells led to unique CD23+ and CD23- subsets that were evident in Mef2c knockout primary samples as well as Mef2c-deficient cultured, differentiated B-cells. Genome-wide expression analysis of immature and mature B-cells lacking Mef2c indicated altered expression for a number of key regulatory proteins for B-cell function including Ciita, CD23, Cr1/Cr2 and Tnfsf4. Chromatin immunoprecipitation analysis confirmed Mef2c binding to the promoters of these genes indicating a direct link between the presence (or absence) of Mef2c and altered transcriptional control in mature B-cells.
Collapse
Affiliation(s)
- Irina Debnath
- The Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | | | | | | | | |
Collapse
|
39
|
Gagan J, Dey BK, Layer R, Yan Z, Dutta A. Notch3 and Mef2c proteins are mutually antagonistic via Mkp1 protein and miR-1/206 microRNAs in differentiating myoblasts. J Biol Chem 2012; 287:40360-70. [PMID: 23055528 DOI: 10.1074/jbc.m112.378414] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Notch3 is expressed in myogenic precursors, but its function is not well known. RESULTS Notch3 represses the activity of Mef2c and is in turn inhibited by the microRNAs-1 and -206. CONCLUSION Notch3 serves as a regulator for preventing premature myogenic differentiation. SIGNIFICANCE Understanding how precocious differentiation is prevented is critical for designing therapy for skeletal muscle regeneration. The Notch signaling pathway is a well known regulator of skeletal muscle stem cells known as satellite cells. Loss of Notch1 signaling leads to spontaneous myogenic differentiation. Notch1, normally expressed in satellite cells, is targeted for proteasomal degradation by Numb during differentiation. A homolog of Notch1, Notch3, is also expressed in these cells but is not inhibited by Numb. We find that Notch3 is paradoxically up-regulated during the early stages of differentiation by an enhancer that requires both MyoD and activated Notch1. Notch3 itself strongly inhibits the myogenic transcription factor Mef2c, most likely by increasing the p38 phosphatase Mkp1, which inhibits the Mef2c activator p38 MAP kinase. Active Notch3 decreases differentiation. Mef2c, however, induces microRNAs miR-1 and miR-206, which directly down-regulate Notch3 and allow differentiation to proceed. Thus, the myogenic differentiation-induced microRNAs miR-1 and -206 are important for differentiation at least partly because they turn off Notch3. We suggest that the transient expression of Notch3 early in differentiation generates a temporal lag between myoblast activation by MyoD and terminal differentiation into myotubes directed by Mef2c.
Collapse
Affiliation(s)
- Jeffrey Gagan
- Department of Biochemistry and Molecular Genetics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
40
|
Pallavi SK, Ho DM, Hicks C, Miele L, Artavanis-Tsakonas S. Notch and Mef2 synergize to promote proliferation and metastasis through JNK signal activation in Drosophila. EMBO J 2012; 31:2895-907. [PMID: 22580825 PMCID: PMC3395089 DOI: 10.1038/emboj.2012.129] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 04/02/2012] [Indexed: 12/31/2022] Open
Abstract
Genetic analyses in Drosophila revealed a synergy between Notch and the pleiotropic transcription factor Mef2 (myocyte enhancer factor 2), which profoundly influences proliferation and metastasis. We show that these hyperproliferative and invasive Drosophila phenotypes are attributed to upregulation of eiger, a member of the tumour necrosis factor superfamily of ligands, and the consequent activation of Jun N-terminal kinase signalling, which in turn triggers the expression of the invasive marker MMP1. Expression studies in human breast tumour samples demonstrate correlation between Notch and Mef2 paralogues and support the notion that Notch-MEF2 synergy may be significant for modulating human mammary oncogenesis.
Collapse
Affiliation(s)
- S K Pallavi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
41
|
Belyea B, Kephart JG, Blum J, Kirsch DG, Linardic CM. Embryonic signaling pathways and rhabdomyosarcoma: contributions to cancer development and opportunities for therapeutic targeting. Sarcoma 2012; 2012:406239. [PMID: 22619564 PMCID: PMC3350847 DOI: 10.1155/2012/406239] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 01/17/2012] [Indexed: 11/18/2022] Open
Abstract
Rhabdomyosarcoma is the most common soft tissue sarcoma of childhood and adolescence, accounting for approximately 7% of childhood cancers. Current therapies include nonspecific cytotoxic chemotherapy regimens, radiation therapy, and surgery; however, these multimodality strategies are unsuccessful in the majority of patients with high-risk disease. It is generally believed that these tumors represent arrested or aberrant skeletal muscle development, and, accordingly, developmental signaling pathways critical to myogenesis such as Notch, WNT, and Hedgehog may represent new therapeutic targets. In this paper, we summarize the current preclinical studies linking these embryonic pathways to rhabdomyosarcoma tumorigenesis and provide support for the investigation of targeted therapies in this embryonic cancer.
Collapse
Affiliation(s)
- Brian Belyea
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA
| | - Julie Grondin Kephart
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jordan Blum
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - David G. Kirsch
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Corinne M. Linardic
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
42
|
Constitutive Notch activation upregulates Pax7 and promotes the self-renewal of skeletal muscle satellite cells. Mol Cell Biol 2012; 32:2300-11. [PMID: 22493066 DOI: 10.1128/mcb.06753-11] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Notch signaling is a conserved cell fate regulator during development and postnatal tissue regeneration. Using skeletal muscle satellite cells as a model and through myogenic cell lineage-specific NICD(OE) (overexpression of constitutively activated Notch 1 intracellular domain), here we investigate how Notch signaling regulates the cell fate choice of muscle stem cells. We show that in addition to inhibiting MyoD and myogenic differentiation, NICD(OE) upregulates Pax7 and promotes the self-renewal of satellite cell-derived primary myoblasts in culture. Using MyoD(-/-) myoblasts, we further show that NICD(OE) upregulates Pax7 independently of MyoD inhibition. In striking contrast to previous observations, NICD(OE) also inhibits S-phase entry and Ki67 expression and thus reduces the proliferation of primary myoblasts. Overexpression of canonical Notch target genes mimics the inhibitory effects of NICD(OE) on MyoD and Ki67 but not the stimulatory effect on Pax7. Instead, NICD regulates Pax7 through interaction with RBP-Jκ, which binds to two consensus sites upstream of the Pax7 gene. Importantly, satellite cell-specific NICD(OE) results in impaired regeneration of skeletal muscles along with increased Pax7(+) mononuclear cells. Our results establish a role of Notch signaling in actively promoting the self-renewal of muscle stem cells through direct regulation of Pax7.
Collapse
|
43
|
Andersen P, Uosaki H, Shenje LT, Kwon C. Non-canonical Notch signaling: emerging role and mechanism. Trends Cell Biol 2012; 22:257-65. [PMID: 22397947 DOI: 10.1016/j.tcb.2012.02.003] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/27/2012] [Accepted: 02/09/2012] [Indexed: 02/08/2023]
Abstract
Notch is an ancient transmembrane receptor with crucial roles in cell-fate choices. Although the 'canonical' Notch pathway and its core members are well established - involving ligand-induced cleavage of Notch for transcriptional regulation - it has been unclear whether Notch can also function independently of ligand and transcription ('non-canonically') through a common mechanism. Recent studies suggest that Notch can non-canonically exert its biological functions by post-translationally targeting Wnt/β-catenin signaling, an important cellular and developmental regulator. The non-canonical Notch pathway appears to be highly conserved from flies to mammals. Here, we discuss the emerging conserved mechanism and role of ligand/transcription-independent Notch signaling in cell and developmental biology.
Collapse
Affiliation(s)
- Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins University, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | | | | | |
Collapse
|
44
|
A long noncoding RNA controls muscle differentiation by functioning as a competing endogenous RNA. Cell 2011; 147:358-69. [PMID: 22000014 PMCID: PMC3234495 DOI: 10.1016/j.cell.2011.09.028] [Citation(s) in RCA: 2160] [Impact Index Per Article: 154.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/08/2011] [Accepted: 09/22/2011] [Indexed: 12/11/2022]
Abstract
Recently, a new regulatory circuitry has been identified in which RNAs can crosstalk with each other by competing for shared microRNAs. Such competing endogenous RNAs (ceRNAs) regulate the distribution of miRNA molecules on their targets and thereby impose an additional level of post-transcriptional regulation. Here we identify a muscle-specific long noncoding RNA, linc-MD1, which governs the time of muscle differentiation by acting as a ceRNA in mouse and human myoblasts. Downregulation or overexpression of linc-MD1 correlate with retardation or anticipation of the muscle differentiation program, respectively. We show that linc-MD1 “sponges” miR-133 and miR-135 to regulate the expression of MAML1 and MEF2C, transcription factors that activate muscle-specific gene expression. Finally, we demonstrate that linc-MD1 exerts the same control over differentiation timing in human myoblasts, and that its levels are strongly reduced in Duchenne muscle cells. We conclude that the ceRNA network plays an important role in muscle differentiation.
Collapse
|
45
|
Beres BJ, George R, Lougher EJ, Barton M, Verrelli BC, McGlade CJ, Rawls JA, Wilson-Rawls J. Numb regulates Notch1, but not Notch3, during myogenesis. Mech Dev 2011; 128:247-57. [DOI: 10.1016/j.mod.2011.02.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 02/18/2011] [Accepted: 02/22/2011] [Indexed: 12/18/2022]
|
46
|
Tee JM, Peppelenbosch MP. Anchoring skeletal muscle development and disease: the role of ankyrin repeat domain containing proteins in muscle physiology. Crit Rev Biochem Mol Biol 2010; 45:318-30. [PMID: 20515317 PMCID: PMC2942773 DOI: 10.3109/10409238.2010.488217] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The ankyrin repeat is a protein module with high affinity for other ankyrin repeats based on strong Van der Waals forces. The resulting dimerization is unusually resistant to both mechanical forces and alkanization, making this module exceedingly useful for meeting the extraordinary demands of muscle physiology. Many aspects of muscle function are controlled by the superfamily ankyrin repeat domain containing proteins, including structural fixation of the contractile apparatus to the muscle membrane by ankyrins, the archetypical member of the family. Additionally, other ankyrin repeat domain containing proteins critically control the various differentiation steps during muscle development, with Notch and developmental stage-specific expression of the members of the Ankyrin repeat and SOCS box (ASB) containing family of proteins controlling compartment size and guiding the various steps of muscle specification. Also, adaptive responses in fully formed muscle require ankyrin repeat containing proteins, with Myotrophin/V-1 ankyrin repeat containing proteins controlling the induction of hypertrophic responses following excessive mechanical load, and muscle ankyrin repeat proteins (MARPs) acting as protective mechanisms of last resort following extreme demands on muscle tissue. Knowledge on mechanisms governing the ordered expression of the various members of superfamily of ankyrin repeat domain containing proteins may prove exceedingly useful for developing novel rational therapy for cardiac disease and muscle dystrophies.
Collapse
Affiliation(s)
- Jin-Ming Tee
- Hubrecht Institute for Developmental Biology and Stem Cell Research-University Medical Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | | |
Collapse
|
47
|
Buas MF, Kadesch T. Regulation of skeletal myogenesis by Notch. Exp Cell Res 2010; 316:3028-33. [PMID: 20452344 DOI: 10.1016/j.yexcr.2010.05.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 05/03/2010] [Indexed: 12/11/2022]
Abstract
Notch signaling has emerged as a key player in skeletal muscle development and regeneration. Simply stated, Notch signaling inhibits differentiation. Accordingly, fine-tuning the pathway is essential for proper muscle homeostasis. This review will address various aspects of Notch signaling, including our current views of the core pathway, its effects in muscle, its interactions with other signaling pathways, and its relationship with ageing.
Collapse
Affiliation(s)
- Matthew F Buas
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6145, USA
| | | |
Collapse
|
48
|
Bakkar N, Guttridge DC. NF-kappaB signaling: a tale of two pathways in skeletal myogenesis. Physiol Rev 2010; 90:495-511. [PMID: 20393192 DOI: 10.1152/physrev.00040.2009] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
NF-kappaB is a ubiquitiously expressed transcription factor that plays vital roles in innate immunity and other processes involving cellular survival, proliferation, and differentiation. Activation of NF-kappaB is controlled by an IkappaB kinase (IKK) complex that can direct either canonical (classical) NF-kappaB signaling by degrading the IkappaB inhibitor and releasing p65/p50 dimers to the nucleus, or causes p100 processing and nuclear translocation of RelB/p52 via a noncanonical (alternative) pathway. Under physiological conditions, NF-kappaB activity is transiently regulated, whereas constitutive activation of this transcription factor typically in the classical pathway is associated with a multitude of disease conditions, including those related to skeletal muscle. How NF-kappaB functions in muscle diseases is currently under intense investigation. Insight into this role of NF-kappaB may be gained by understanding at a more basic level how this transcription factor contributes to skeletal muscle cell differentiation. Recent data from knockout mice support that the classical NF-kappaB pathway functions as an inhibitor of skeletal myogenesis and muscle regeneration acting through multiple mechanisms. In contrast, alternative NF-kappaB signaling does not appear to be required for myofiber conversion, but instead functions in myotube homeostasis by regulating mitochondrial biogenesis. Additional knowledge of these signaling pathways in skeletal myogenesis should aid in the development of specific inhibitors that may be useful in treatments of muscle disorders.
Collapse
Affiliation(s)
- Nadine Bakkar
- Department of Molecular Virology, Immunology, and Medical Genetics, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
49
|
Abstract
The Notch-signaling pathway is involved in multiple processes during vertebrate cardiac development. Cardiomyocyte differentiation, patterning of the different cardiac regions, valve development, ventricular trabeculation, and outflow tract development have all been shown to depend on the activity of specific Notch-signaling elements. From these studies, it becomes obvious that Notch regulates in a cell autonomous or non-cell autonomous manner different signaling pathways, pointing to a role for Notch as a signal coordinator during cardiogenesis. While most of the research has concentrated on Notch signaling in the myocardium, the importance of Notch activity in the cardiac endothelium (endocardium) must not be overlooked. Endocardial Notch activity is crucial for valve and ventricular trabeculae development, two processes that illustrate the role of Notch as a signal coordinator. The importance of Notch signaling in human disease is evident from the discovery that many mutations in components of this pathway segregate in several inherited and acquired disorders. This reflects the fundamental roles that Notch performs during cardiac ontogeny. This review examines the experimental evidence supporting a role for Notch in cardiac development and adult heart homeostasis, and how dysregulated Notch signaling may lead to cardiac disease in the newborn and in the adult.
Collapse
|
50
|
Gang EJ, Darabi R, Bosnakovski D, Xu Z, Kamm KE, Kyba M, Perlingeiro RCR. Engraftment of mesenchymal stem cells into dystrophin-deficient mice is not accompanied by functional recovery. Exp Cell Res 2009; 315:2624-36. [PMID: 19460366 DOI: 10.1016/j.yexcr.2009.05.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 05/05/2009] [Accepted: 05/06/2009] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cell preparations have been proposed for muscle regeneration in musculoskeletal disorders. Although MSCs have great in vitro expansion potential and possess the ability to differentiate into several mesenchymal lineages, myogenesis has proven to be much more difficult to induce. We have recently demonstrated that Pax3, the master regulator of the embryonic myogenic program, enables the in vitro differentiation of a murine mesenchymal stem cell line (MSCB9-Pax3) into myogenic progenitors. Here we show that injection of these cells into cardiotoxin-injured muscles of immunodeficient mice leads to the development of muscle tumors, resembling rhabdomyosarcomas. We then extended these studies to primary human mesenchymal stem cells (hMSCs) isolated from bone marrow. Upon genetic modification with a lentiviral vector encoding PAX3, hMSCs activated the myogenic program as demonstrated by expression of myogenic regulatory factors. Upon transplantation, the PAX3-modified MSCs did not generate rhabdomyosarcomas but rather, resulted in donor-derived myofibers. These were found at higher frequency in PAX3-transduced hMSCs than in mock-transduced MSCs. Nonetheless, neither engraftment of PAX3-modified or unmodified MSCs resulted in improved contractility. Thus these findings suggest that limitations remain to be overcome before MSC preparations result in effective treatment for muscular dystrophies.
Collapse
Affiliation(s)
- Eun Ji Gang
- Department of Developmental Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9133, USA
| | | | | | | | | | | | | |
Collapse
|