151
|
Zhou J, So KK, Li Y, Li Y, Yuan J, Ding Y, Chen F, Huang Y, Liu J, Lee W, Li G, Ju Z, Sun H, Wang H. Elevated H3K27ac in aged skeletal muscle leads to increase in extracellular matrix and fibrogenic conversion of muscle satellite cells. Aging Cell 2019; 18:e12996. [PMID: 31325224 PMCID: PMC6718601 DOI: 10.1111/acel.12996] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 05/24/2019] [Accepted: 06/05/2019] [Indexed: 01/22/2023] Open
Abstract
Epigenetic alterations occur in various cells and tissues during aging, but it is not known if such alterations are also associated with aging in skeletal muscle. Here, we examined the changes of a panel of histone modifications and found H3K27ac (an active enhancer mark) is markedly increased in aged human skeletal muscle tissues. Further analyses uncovered that the H3K27ac increase and enhancer activation are associated with the up‐regulation of extracellular matrix (ECM) genes; this may result in alteration of the niche environment for skeletal muscle stem cells, also called satellite cells (SCs), which causes decreased myogenic potential and fibrogenic conversion of SCs. In mice, treatment of aging muscles with JQ1, an inhibitor of enhancer activation, inhibited the ECM up‐regulation and fibrogenic conversion of SCs and restored their myogenic differentiation potential. Altogether, our findings not only uncovered a novel aspect of skeletal muscle aging that is associated with enhancer remodeling but also implicated JQ1 as a potential treatment approach for restoring SC function in aging muscle.
Collapse
Affiliation(s)
- Jiajian Zhou
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Karl K. So
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Yuying Li
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Yang Li
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Department of Orthopaedics and Traumatology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Jie Yuan
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Yingzhe Ding
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Fengyuan Chen
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Yile Huang
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Jin Liu
- Key Laboratory of Regenerative Medicine of Ministry of Education Institute of Aging and Regenerative MedicineJinan University Guangzhou China
| | - Wayne Lee
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Department of Orthopaedics and Traumatology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Gang Li
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Department of Orthopaedics and Traumatology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education Institute of Aging and Regenerative MedicineJinan University Guangzhou China
| | - Hao Sun
- Department of Chemical Pathology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| | - Huating Wang
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
- Department of Orthopaedics and Traumatology The Chinese University of Hong Kong, Prince of Wales Hospital Shatin, New Territories Hong Kong SAR China
| |
Collapse
|
152
|
Huang R, Zong X, Nadesan P, Huebner JL, Kraus VB, White JP, White PJ, Baht GS. Lowering circulating apolipoprotein E levels improves aged bone fracture healing. JCI Insight 2019; 4:129144. [PMID: 31534056 DOI: 10.1172/jci.insight.129144] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/08/2019] [Indexed: 01/22/2023] Open
Abstract
Age is a well-established risk factor for impaired bone fracture healing. Here, we identify a role for apolipoprotein E (ApoE) in age-associated impairment of bone fracture healing and osteoblast differentiation, and we investigate the mechanism by which ApoE alters these processes. We identified that, in both humans and mice, circulating ApoE levels increase with age. We assessed bone healing in WT and ApoE-/- mice after performing tibial fracture surgery: bone deposition was higher within fracture calluses from ApoE-/- mice. In vitro recombinant ApoE (rApoE) treatment of differentiating osteoblasts decreased cellular differentiation and matrix mineralization. Moreover, this rApoE treatment decreased osteoblast glycolytic activity while increasing lipid uptake and fatty acid oxidation. Using parabiosis models, we determined that circulating ApoE plays a strong inhibitory role in bone repair. Using an adeno-associated virus-based siRNA system, we decreased circulating ApoE levels in 24-month-old mice and demonstrated that, as a result, fracture calluses from these aged mice displayed enhanced bone deposition and mechanical strength. Our results demonstrate that circulating ApoE as an aging factor inhibits bone fracture healing by altering osteoblast metabolism, thereby identifying ApoE as a new therapeutic target for improving bone repair in the elderly.
Collapse
Affiliation(s)
- Rong Huang
- Duke Molecular Physiology Institute.,Department of Orthopaedic Surgery
| | - Xiaohua Zong
- Duke Molecular Physiology Institute.,Department of Orthopaedic Surgery
| | | | | | - Virginia B Kraus
- Duke Molecular Physiology Institute.,Department of Orthopaedic Surgery.,Department of Pathology, and.,Department of Medicine, Duke University, Durham, North Carolina, USA
| | - James P White
- Duke Molecular Physiology Institute.,Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Phillip J White
- Duke Molecular Physiology Institute.,Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Gurpreet S Baht
- Duke Molecular Physiology Institute.,Department of Orthopaedic Surgery.,Department of Pathology, and
| |
Collapse
|
153
|
Forte E, Furtado MB, Rosenthal N. The interstitium in cardiac repair: role of the immune-stromal cell interplay. Nat Rev Cardiol 2019; 15:601-616. [PMID: 30181596 DOI: 10.1038/s41569-018-0077-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiac regeneration, that is, restoration of the original structure and function in a damaged heart, differs from tissue repair, in which collagen deposition and scar formation often lead to functional impairment. In both scenarios, the early-onset inflammatory response is essential to clear damaged cardiac cells and initiate organ repair, but the quality and extent of the immune response vary. Immune cells embedded in the damaged heart tissue sense and modulate inflammation through a dynamic interplay with stromal cells in the cardiac interstitium, which either leads to recapitulation of cardiac morphology by rebuilding functional scaffolds to support muscle regrowth in regenerative organisms or fails to resolve the inflammatory response and produces fibrotic scar tissue in adult mammals. Current investigation into the mechanistic basis of homeostasis and restoration of cardiac function has increasingly shifted focus away from stem cell-mediated cardiac repair towards a dynamic interplay of cells composing the less-studied interstitial compartment of the heart, offering unexpected insights into the immunoregulatory functions of cardiac interstitial components and the complex network of cell interactions that must be considered for clinical intervention in heart diseases.
Collapse
Affiliation(s)
| | | | - Nadia Rosenthal
- The Jackson Laboratory, Bar Harbor, ME, USA. .,National Heart and Lung Institute, Imperial College London, Faculty of Medicine, Imperial Centre for Translational and Experimental Medicine, London, UK.
| |
Collapse
|
154
|
Farzaneh M, Alishahi M, Derakhshan Z, Sarani NH, Attari F, Khoshnam SE. The Expression and Functional Roles of miRNAs in Embryonic and Lineage-Specific Stem Cells. Curr Stem Cell Res Ther 2019; 14:278-289. [PMID: 30674265 DOI: 10.2174/1574888x14666190123162402] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/15/2018] [Accepted: 01/03/2019] [Indexed: 01/01/2023]
Abstract
The discovery of small non-coding RNAs began an interesting era in cellular and molecular biology. To date, miRNAs are the best recognized non-coding RNAs for maintenance and differentiation of pluripotent stem cells including embryonic stem cells (ES), induced pluripotent stem cells (iPSC), and cancer stem cells. ES cells are defined by their ability to self-renew, teratoma formation, and to produce numerous types of differentiated cells. Dual capacity of ES cells for self-renewal and differentiation is controlled by specific interaction with the neighboring cells and intrinsic signaling pathways from the level of transcription to translation. The ES cells have been the suitable model for evaluating the function of non-coding RNAs and in specific miRNAs. So far, the general function of the miRNAs in ES cells has been assessed in mammalian and non-mammalian stem cells. Nowadays, the evolution of sequencing technology led to the discovery of numerous miRNAs in human and mouse ES cells that their expression levels significantly changes during proliferation and differentiation. Several miRNAs have been identified in ectoderm, mesoderm, and endoderm cells, as well. This review would focus on recent knowledge about the expression and functional roles of miRNAs in embryonic and lineage-specific stem cells. It also describes that miRNAs might have essential roles in orchestrating the Waddington's landscape structure during development.
Collapse
Affiliation(s)
- Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masoumeh Alishahi
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Derakhshan
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda H Sarani
- Faculty of Paramedical, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnoosh Attari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Seyed E Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
155
|
Direct Reprogramming of Mouse Fibroblasts into Functional Skeletal Muscle Progenitors. Stem Cell Reports 2019; 10:1505-1521. [PMID: 29742392 PMCID: PMC5995754 DOI: 10.1016/j.stemcr.2018.04.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle harbors quiescent stem cells termed satellite cells and proliferative progenitors termed myoblasts, which play pivotal roles during muscle regeneration. However, current technology does not allow permanent capture of these cell populations in vitro. Here, we show that ectopic expression of the myogenic transcription factor MyoD, combined with exposure to small molecules, reprograms mouse fibroblasts into expandable induced myogenic progenitor cells (iMPCs). iMPCs express key skeletal muscle stem and progenitor cell markers including Pax7 and Myf5 and give rise to dystrophin-expressing myofibers upon transplantation in vivo. Notably, a subset of transplanted iMPCs maintain Pax7 expression and sustain serial regenerative responses. Similar to satellite cells, iMPCs originate from Pax7+ cells and require Pax7 itself for maintenance. Finally, we show that myogenic progenitor cell lines can be established from muscle tissue following small-molecule exposure alone. This study thus reports on a robust approach to derive expandable myogenic stem/progenitor-like cells from multiple cell types. MyoD and small molecules reprogram fibroblasts to myogenic progenitors termed iMPCs iMPCs self-renew and express key satellite cell and myoblast markers iMPC growth is driven by Pax7+ cells and requires Pax7 gene function Transplanted iMPCs engraft and sustain muscle regeneration in vivo
Collapse
|
156
|
Fate decision of satellite cell differentiation and self-renewal by miR-31-IL34 axis. Cell Death Differ 2019; 27:949-965. [PMID: 31332295 DOI: 10.1038/s41418-019-0390-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 06/13/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022] Open
Abstract
Quiescent satellite cells (SCs) that are activated to produce numerous myoblasts underpin the complete healing of damaged skeletal muscle. How cell-autonomous regulatory mechanisms modulate the balance among cells committed to differentiation and those committed to self-renewal to maintain the stem cell pool remains poorly explored. Here, we show that miR-31 inactivation compromises muscle regeneration in adult mice by impairing the expansion of myoblasts. miR-31 is pivotal for SC proliferation, and its deletion promotes asymmetric cell fate segregation of proliferating cells, resulting in enhanced myogenic commitment and re-entry into quiescence. Further analysis revealed that miR-31 posttranscriptionally suppresses interleukin 34 (IL34) mRNA, the protein product of which activates JAK-STAT3 signaling required for myogenic progression. IL34 inhibition rescues the regenerative deficiency of miR-31 knockout mice. Our results provide evidence that targeting miR-31 or IL34 activities in SCs could be used to counteract the functional exhaustion of SCs in pathological conditions.
Collapse
|
157
|
A novel mitochondrial micropeptide MPM enhances mitochondrial respiratory activity and promotes myogenic differentiation. Cell Death Dis 2019; 10:528. [PMID: 31296841 PMCID: PMC6624212 DOI: 10.1038/s41419-019-1767-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/25/2019] [Accepted: 06/26/2019] [Indexed: 12/14/2022]
Abstract
Micropeptides belong to a class of newly identified small molecules with <100 amino acids in length, and their functions remain largely unknown. Here, we identified a novel muscle-enriched micropeptide that was localized to mitochondria (named MPM, micropeptide in mitochondria) and upregulated during in vitro differentiation of C2C12 myoblasts and in vivo early postnatal skeletal muscle development, and muscle regeneration after cardiotoxin (CTX) damage. Downregulation of MPM was observed in the muscular tissues of tibial muscular dystrophy and Duchenne muscular dystrophy patients. Furthermore, MPM silencing inhibited the differentiation of C2C12 myoblasts into myotubes, whereas MPM overexpression stimulated it. MPM−/− mice exhibited smaller skeletal muscle fibers and worse muscle performance, such as decrease in the maximum grip force of limbs, the latency to fall off rotarod, and the exhausting swimming time. Muscle regeneration was also impaired in MPM−/− mice, as evidenced by lower expression of Pax7, MyoD, and MyoG after CTX injection and smaller regenerated myofibers, compared with wild-type mice. Mechanistical investigations based on both gain- and loss-of function studies revealed that MPM increased oxygen consumption and ATP production of mitochondria. Moreover, ectopic expression of PGC-1α, which can enhance mitochondrial respiration, attenuated the inhibitory effect of siMPM on myogenic differentiation. These results imply that MPM may promote myogenic differentiation and muscle fiber growth by enhancing mitochondrial respiratory activity, which highlights the importance of micropeptides in the elaborate regulatory network of both myogenesis and mitochondrial activity and implicates MPM as a potential target for muscular dystrophy therapy.
Collapse
|
158
|
Schaaf GJ, Canibano-Fraile R, van Gestel TJM, van der Ploeg AT, Pijnappel WWMP. Restoring the regenerative balance in neuromuscular disorders: satellite cell activation as therapeutic target in Pompe disease. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:280. [PMID: 31392192 DOI: 10.21037/atm.2019.04.48] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Skeletal muscle is capable of efficiently regenerating after damage in a process mediated by tissue-resident stem cells called satellite cells. This regenerative potential is often compromised under muscle-degenerative conditions. Consequently, the damage produced during degeneration is not efficiently repaired and the balance between repair and damage is lost. Here we review recent progress on the role of satellite cell-mediated repair in neuromuscular disorders with a focus on Pompe disease, an inherited metabolic myopathy caused by deficiency of the lysosomal enzyme acid alpha glucosidase (GAA). Studies performed in patient biopsies as well as in Pompe disease mouse models demonstrate that muscle regeneration activity is compromised despite progressing muscle damage. We describe disease-specific mechanisms of satellite cell dysfunction to highlight the differences between Pompe disease and muscle dystrophies. The mechanisms involved provide possible targets for therapy, such as modulation of autophagy, muscle exercise, and pharmacological modulation of satellite cell activation. Most of these approaches are still experimental, although promising in animal models, still warrant caution with respect to their safety and efficiency profile.
Collapse
Affiliation(s)
- Gerben J Schaaf
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rodrigo Canibano-Fraile
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Tom J M van Gestel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ans T van der Ploeg
- Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - W W M Pim Pijnappel
- Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
159
|
Miller BM, Johnson K, Whited JL. Common themes in tetrapod appendage regeneration: a cellular perspective. EvoDevo 2019; 10:11. [PMID: 31236203 PMCID: PMC6572735 DOI: 10.1186/s13227-019-0124-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/08/2019] [Indexed: 01/13/2023] Open
Abstract
Complete and perfect regeneration of appendages is a process that has fascinated and perplexed biologists for centuries. Some tetrapods possess amazing regenerative abilities, but the regenerative abilities of others are exceedingly limited. The reasons underlying these differences have largely remained mysterious. A great deal has been learned about the morphological events that accompany successful appendage regeneration, and a handful of experimental manipulations can be reliably applied to block the process. However, only in the last decade has the goal of attaining a thorough molecular and cellular biological understanding of appendage regeneration in tetrapods become within reach. Advances in molecular and genetic tools for interrogating these remarkable events are now allowing for unprecedented access to the fundamental biology at work in appendage regeneration in a variety of species. This information will be critical for integrating the large body of detailed observations from previous centuries with a modern understanding of how cells sense and respond to severe injury and loss of body parts. Understanding commonalities between regenerative modes across diverse species is likely to illuminate the most important aspects of complex tissue regeneration.
Collapse
Affiliation(s)
- Bess M. Miller
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave, Cambridge, MA 02138 USA
| | - Kimberly Johnson
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave, Cambridge, MA 02138 USA
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, 7 Divinity Ave, Cambridge, MA 02138 USA
| |
Collapse
|
160
|
Ghanim H, Dhindsa S, Batra M, Green K, Abuaysheh S, Kuhadiya ND, Makdissi A, Chaudhuri A, Dandona P. Effect of Testosterone on FGF2, MRF4, and Myostatin in Hypogonadotropic Hypogonadism: Relevance to Muscle Growth. J Clin Endocrinol Metab 2019; 104:2094-2102. [PMID: 30629183 PMCID: PMC6481910 DOI: 10.1210/jc.2018-01832] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022]
Abstract
CONTEXT Fibroblast growth factor (FGF)2 is an important stimulatory modulator of satellite cells in skeletal muscle. Satellite cells play a cardinal role in muscle growth and repair. OBJECTIVE We evaluated whether skeletal muscle expression of FGF2 and muscle growth and differentiation factors are reduced in patients with hypogonadotropic hypogonadism (HH) and whether testosterone replacement therapy results in their restoration. DESIGN This is a secondary analysis of a previously completed trial of testosterone replacement in men with type 2 diabetes and HH. SETTING Clinical Research Center at a university. PATIENTS Twenty-two men with HH and 20 eugonadal men were compared at baseline. INTERVENTIONS Twelve men with HH were treated with intramuscular injections of 250 mg testosterone every 2 weeks for 22 weeks, and 10 men received placebo injections. Quadriceps muscle biopsies and blood samples were obtained before and after testosterone therapy. OUTCOME MEASURES AND RESULTS The expression of FGF2 and FGF receptor (FGFR)2 in skeletal muscle of men with HH was significantly lower than that in eugonadal men by 57% and 39%, respectively (P < 0.05). After 22 weeks of testosterone, the expression of FGF2 increased, whereas that of myogenic regulatory factor (MRF)4 and myostatin decreased significantly. There was no change in expression of FGFR2, myogenin, or myogenic differentiation protein in the skeletal muscle. Plasma FGF2 and IGF-1 concentrations increased after testosterone therapy. CONCLUSIONS These data show that testosterone is a major modulator of FGF2, MRF4, and myostatin expression in skeletal muscle. These effects may contribute to the increase in muscle mass after testosterone therapy.
Collapse
Affiliation(s)
- Husam Ghanim
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
| | - Sandeep Dhindsa
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
- Division of Endocrinology, Diabetes and Metabolism, Saint Louis University, St. Louis, Missouri 63104
| | - Manav Batra
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
| | - Kelly Green
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
| | - Sanaa Abuaysheh
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
| | - Nitesh D Kuhadiya
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
| | - Antoine Makdissi
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
| | - Ajay Chaudhuri
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
| | - Paresh Dandona
- Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, Williamsville, New York 14221
- Correspondence and Reprint Requests: Paresh Dandona, MD, PhD, Division of Endocrinology, Diabetes and Metabolism, State University of New York at Buffalo, 1000 Youngs Road, Suite 105, Williamsville, New York 14221. E-mail:
| |
Collapse
|
161
|
Mohammad K, Dakik P, Medkour Y, Mitrofanova D, Titorenko VI. Quiescence Entry, Maintenance, and Exit in Adult Stem Cells. Int J Mol Sci 2019; 20:ijms20092158. [PMID: 31052375 PMCID: PMC6539837 DOI: 10.3390/ijms20092158] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 12/13/2022] Open
Abstract
Cells of unicellular and multicellular eukaryotes can respond to certain environmental cues by arresting the cell cycle and entering a reversible state of quiescence. Quiescent cells do not divide, but can re-enter the cell cycle and resume proliferation if exposed to some signals from the environment. Quiescent cells in mammals and humans include adult stem cells. These cells exhibit improved stress resistance and enhanced survival ability. In response to certain extrinsic signals, adult stem cells can self-renew by dividing asymmetrically. Such asymmetric divisions not only allow the maintenance of a population of quiescent cells, but also yield daughter progenitor cells. A multistep process of the controlled proliferation of these progenitor cells leads to the formation of one or more types of fully differentiated cells. An age-related decline in the ability of adult stem cells to balance quiescence maintenance and regulated proliferation has been implicated in many aging-associated diseases. In this review, we describe many traits shared by different types of quiescent adult stem cells. We discuss how these traits contribute to the quiescence, self-renewal, and proliferation of adult stem cells. We examine the cell-intrinsic mechanisms that allow establishing and sustaining the characteristic traits of adult stem cells, thereby regulating quiescence entry, maintenance, and exit.
Collapse
Affiliation(s)
- Karamat Mohammad
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Paméla Dakik
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Younes Medkour
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Darya Mitrofanova
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| | - Vladimir I Titorenko
- Department of Biology, Concordia University, 7141 Sherbrooke Street, West, SP Building, Room 501-13, Montreal, QC H4B 1R6, Canada.
| |
Collapse
|
162
|
Banerji CRS, Panamarova M, Pruller J, Figeac N, Hebaishi H, Fidanis E, Saxena A, Contet J, Sacconi S, Severini S, Zammit PS. Dynamic transcriptomic analysis reveals suppression of PGC1α/ERRα drives perturbed myogenesis in facioscapulohumeral muscular dystrophy. Hum Mol Genet 2019; 28:1244-1259. [PMID: 30462217 PMCID: PMC6452176 DOI: 10.1093/hmg/ddy405] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 01/06/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a prevalent, incurable myopathy, linked to epigenetic derepression of D4Z4 repeats on chromosome 4q, leading to ectopic DUX4 expression. FSHD patient myoblasts have defective myogenic differentiation, forming smaller myotubes with reduced myosin content. However, molecular mechanisms driving such disrupted myogenesis in FSHD are poorly understood. We performed high-throughput morphological analysis describing FSHD and control myogenesis, revealing altered myogenic differentiation results in hypotrophic myotubes. Employing polynomial models and an empirical Bayes approach, we established eight critical time points during which human healthy and FSHD myogenesis differ. RNA-sequencing at these eight nodal time points in triplicate, provided temporal depth for a multivariate regression analysis, allowing assessment of interaction between progression of differentiation and FSHD disease status. Importantly, the unique size and structure of our data permitted identification of many novel FSHD pathomechanisms undetectable by previous approaches. For further analysis here, we selected pathways that control mitochondria: of interest considering known alterations in mitochondrial structure and function in FSHD muscle, and sensitivity of FSHD cells to oxidative stress. Notably, we identified suppression of mitochondrial biogenesis, in particular via peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC1α), the cofactor and activator of oestrogen-related receptor α (ERRα). PGC1α knock-down caused hypotrophic myotubes to form from control myoblasts. Known ERRα agonists and safe food supplements biochanin A, daidzein or genistein, each rescued the hypotrophic FSHD myotube phenotype. Together our work describes transcriptomic changes in high resolution that occur during myogenesis in FSHD ex vivo, identifying suppression of the PGC1α-ERRα axis leading to perturbed myogenic differentiation, which can effectively be rescued by readily available food supplements.
Collapse
Affiliation(s)
- Christopher R S Banerji
- Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London, UK
- Department of Computer Science, University College London, London, UK
- Centre of Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London, UK
| | - Maryna Panamarova
- Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London, UK
| | - Johanna Pruller
- Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London, UK
| | - Nicolas Figeac
- Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London, UK
| | - Husam Hebaishi
- Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London, UK
| | - Efthymios Fidanis
- Genomics Research Platform, Biomedical Research Centre at Guy’s and St Thomas’ Trust and Kings College London, Guy’s Hospital, London, UK
| | - Alka Saxena
- Genomics Research Platform, Biomedical Research Centre at Guy’s and St Thomas’ Trust and Kings College London, Guy’s Hospital, London, UK
| | - Julian Contet
- Institute for Research on Cancer and Aging of Nice, Faculty of Medicine, Université Côte d'Azur, Nice, Cedex, France
| | - Sabrina Sacconi
- Institute for Research on Cancer and Aging of Nice, Faculty of Medicine, Université Côte d'Azur, Nice, Cedex, France
- Peripheral Nervous System, Muscle and ALS Department, Université Côte d'Azur, Nice, France
| | - Simone Severini
- Department of Computer Science, University College London, London, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London, UK
| |
Collapse
|
163
|
Chen B, Shan T. The role of satellite and other functional cell types in muscle repair and regeneration. J Muscle Res Cell Motil 2019; 40:1-8. [PMID: 30968305 DOI: 10.1007/s10974-019-09511-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Abstract
Skeletal muscles play essential roles in physiological processes, including motor function, energy hemostasis, and respiration. Skeletal muscles also have the capacity to regenerate after injury. Regeneration of skeletal muscle is an extremely complex biological process, which involves multiple cell types. Skeletal muscle stem cells (also known as satellite cells; SCs) are crucial for the development, growth, maintenance and repair of the skeletal muscle. Cell fates and function have been extensively studied in the context of skeletal muscle regeneration. In addition to SCs, other cell types, such as fibro-adipogenic precursors (FAPs), endothelial cells, fibroblasts, pericytes and certain immune cells, play important regulatory roles during skeletal muscle regeneration. In this review, we summarize and discuss the current research progress on the different cell types and their respective functions in skeletal muscle regeneration and repair.
Collapse
Affiliation(s)
- Bide Chen
- College of Animal Sciences, Zhejiang University; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education; Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University; The Key Laboratory of Molecular Animal Nutrition, Ministry of Education; Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China.
| |
Collapse
|
164
|
Naimo MA, Rader EP, Ensey J, Kashon ML, Baker BA. Reduced frequency of resistance-type exercise training promotes adaptation of the aged skeletal muscle microenvironment. J Appl Physiol (1985) 2019; 126:1074-1087. [PMID: 30676867 DOI: 10.1152/japplphysiol.00582.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The purpose of this study was to characterize the growth and remodeling molecular signaling response in aged skeletal muscle following 1 mo of "resistance-type exercise" training. Male Fischer 344 × Brown Norway hybrid rats aged 3 (young) and 30 mo (old) underwent stretch-shortening contraction (SSC) loading 2 or 3 days/wk; muscles were removed 72 h posttraining. Young rats SSC loaded 3 (Y3x) or 2 days/wk (Y2x) adapted via increased work performance. Old rats SSC loaded 3 days/wk (O3x) maladapted via decreased negative work; however, old rats SSC loaded 2 days/wk (O2x) adapted through improved negative and positive work. Y3x, Y2x, and O2x, but not O3x, displayed hypertrophy via larger fiber area and myonuclear domains. Y3x, Y2x, and O2x differentially expressed 19, 30, and 8 phosphatidylinositol 3-kinase-Akt genes, respectively, whereas O3x only expressed 2. Bioinformatics analysis revealed that rats in the adapting groups presented growth and remodeling processes (i.e., increased protein synthesis), whereas O3x demonstrated inflammatory signaling. In conclusion, reducing SSC-loading frequency in aged rodents positively influences the molecular signaling microenvironment, promoting muscle adaptation. NEW & NOTEWORTHY Decreasing resistance-type exercise training frequency in old rodents led to adaptation through enhancements in performance, fiber areas, and myonuclear domains. Modifying frequency influenced the molecular environment through improvements in phosphatidylinositol 3-kinase-Akt pathway-specific expression and bioinformatics indicating increased protein synthesis. Reducing training frequency may be appropriate in older individuals who respond unfavorably to higher frequencies (i.e., maladaptation); overall, modifying the parameters of the exercise prescription can affect the cellular environment, ultimately leading to adaptive or maladaptive outcomes.
Collapse
Affiliation(s)
- Marshall A Naimo
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention , Morgantown, West Virginia.,Division of Exercise Physiology, School of Medicine, West Virginia University , Morgantown, West Virginia
| | - Erik P Rader
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention , Morgantown, West Virginia
| | - James Ensey
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention , Morgantown, West Virginia
| | - Michael L Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention , Morgantown, West Virginia
| | - Brent A Baker
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention , Morgantown, West Virginia
| |
Collapse
|
165
|
Siles L, Ninfali C, Cortés M, Darling DS, Postigo A. ZEB1 protects skeletal muscle from damage and is required for its regeneration. Nat Commun 2019; 10:1364. [PMID: 30910999 PMCID: PMC6434033 DOI: 10.1038/s41467-019-08983-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/12/2019] [Indexed: 12/31/2022] Open
Abstract
The mechanisms linking muscle injury and regeneration are not fully understood. Here we report an unexpected role for ZEB1 regulating inflammatory and repair responses in dystrophic and acutely injured muscles. ZEB1 is upregulated in the undamaged and regenerating myofibers of injured muscles. Compared to wild-type counterparts, Zeb1-deficient injured muscles exhibit enhanced damage that corresponds with a retarded p38-MAPK-dependent transition of their macrophages towards an anti-inflammatory phenotype. Zeb1-deficient injured muscles also display a delayed and poorer regeneration that is accounted by the retarded anti-inflammatory macrophage transition and their intrinsically deficient muscle satellite cells (MuSCs). Macrophages in Zeb1-deficient injured muscles show lower phosphorylation of p38 and its forced activation reverts the enhanced muscle damage and poorer regeneration. MuSCs require ZEB1 to maintain their quiescence, prevent their premature activation following injury, and drive efficient regeneration in dystrophic muscles. These data indicate that ZEB1 protects muscle from damage and is required for its regeneration. Following muscle damage, an inflammatory response is associated to activation of satellite cells, which drive muscle repair. Here, the authors show that upregulation of Zeb1 in macrophages and muscle fibres regulates inflammation, and also show a role for Zeb1 in maintenance of satellite cell quiescence.
Collapse
Affiliation(s)
- Laura Siles
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain
| | - Chiara Ninfali
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain
| | - Marlies Cortés
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain
| | - Douglas S Darling
- Center for Genetics and Molecular Medicine and Department of Immunology and Infectious Diseases, University of Louisville, Louisville, KY, 40202, USA
| | - Antonio Postigo
- Group of Transcriptional Regulation of Gene Expression, Department of Oncology and Hematology, IDIBAPS, 08036, Barcelona, Spain. .,Molecular Targets Program, James G. Brown Cancer Center, Louisville, KY, 40202, USA. .,ICREA, Barcelona, 08010, Spain.
| |
Collapse
|
166
|
Chuang MC, Lin SS, Ohniwa RL, Lee GH, Su YA, Chang YC, Tang MJ, Liu YW. Tks5 and Dynamin-2 enhance actin bundle rigidity in invadosomes to promote myoblast fusion. J Cell Biol 2019; 218:1670-1685. [PMID: 30894403 PMCID: PMC6504888 DOI: 10.1083/jcb.201809161] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/22/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle development requires the cell-cell fusion of differentiated myoblasts to form muscle fibers. The actin cytoskeleton is known to be the main driving force for myoblast fusion; however, how actin is organized to direct intercellular fusion remains unclear. Here we show that an actin- and dynamin-2-enriched protrusive structure, the invadosome, is required for the fusion process of myogenesis. Upon differentiation, myoblasts acquire the ability to form invadosomes through isoform switching of a critical invadosome scaffold protein, Tks5. Tks5 directly interacts with and recruits dynamin-2 to the invadosome and regulates its assembly around actin filaments to strengthen the stiffness of dynamin-actin bundles and invadosomes. These findings provide a mechanistic framework for the acquisition of myogenic fusion machinery during myogenesis and reveal a novel structural function for Tks5 and dynamin-2 in organizing actin filaments in the invadosome to drive membrane fusion.
Collapse
Affiliation(s)
- Mei-Chun Chuang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shan-Shan Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ryosuke L Ohniwa
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Center for Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Gang-Hui Lee
- International Center of Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - You-An Su
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Chang
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Jer Tang
- International Center of Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan.,Department of Physiology, Medical College, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan .,Center of Precision Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
167
|
Larsson L, Degens H, Li M, Salviati L, Lee YI, Thompson W, Kirkland JL, Sandri M. Sarcopenia: Aging-Related Loss of Muscle Mass and Function. Physiol Rev 2019; 99:427-511. [PMID: 30427277 DOI: 10.1152/physrev.00061.2017] [Citation(s) in RCA: 783] [Impact Index Per Article: 156.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sarcopenia is a loss of muscle mass and function in the elderly that reduces mobility, diminishes quality of life, and can lead to fall-related injuries, which require costly hospitalization and extended rehabilitation. This review focuses on the aging-related structural changes and mechanisms at cellular and subcellular levels underlying changes in the individual motor unit: specifically, the perikaryon of the α-motoneuron, its neuromuscular junction(s), and the muscle fibers that it innervates. Loss of muscle mass with aging, which is largely due to the progressive loss of motoneurons, is associated with reduced muscle fiber number and size. Muscle function progressively declines because motoneuron loss is not adequately compensated by reinnervation of muscle fibers by the remaining motoneurons. At the intracellular level, key factors are qualitative changes in posttranslational modifications of muscle proteins and the loss of coordinated control between contractile, mitochondrial, and sarcoplasmic reticulum protein expression. Quantitative and qualitative changes in skeletal muscle during the process of aging also have been implicated in the pathogenesis of acquired and hereditary neuromuscular disorders. In experimental models, specific intervention strategies have shown encouraging results on limiting deterioration of motor unit structure and function under conditions of impaired innervation. Translated to the clinic, if these or similar interventions, by saving muscle and improving mobility, could help alleviate sarcopenia in the elderly, there would be both great humanitarian benefits and large cost savings for health care systems.
Collapse
Affiliation(s)
- Lars Larsson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Hans Degens
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Meishan Li
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Leonardo Salviati
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Young Il Lee
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Wesley Thompson
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - James L Kirkland
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| | - Marco Sandri
- Department of Physiology and Pharmacology, Basic and Clinical Muscle Biology Group, Karolinska Institutet , Stockholm , Sweden ; Section of Clinical Neurophysiology, Department of Clinical Neuroscience, Karolinska Institutet , Stockholm , Sweden ; Department of Biobehavioral Health, The Pennsylvania State University , University Park, Pennsylvania ; School of Healthcare Science, Metropolitan University , Manchester , United Kingdom ; Institute of Sport Science and Innovations, Lithuanian Sports University , Kaunas , Lithuania ; Clinical Genetics Unit, Department of Woman and Child Health, University of Padova , Padova , Italy ; IRP Città della Speranza, Padova , Italy ; Department of Biology, Texas A&M University , College Station, Texas ; Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester, Minnesota ; Department of Biomedical Science, Venetian Institute of Molecular Medicine, University of Padova , Padova , Italy
| |
Collapse
|
168
|
Lukjanenko L, Karaz S, Stuelsatz P, Gurriaran-Rodriguez U, Michaud J, Dammone G, Sizzano F, Mashinchian O, Ancel S, Migliavacca E, Liot S, Jacot G, Metairon S, Raymond F, Descombes P, Palini A, Chazaud B, Rudnicki MA, Bentzinger CF, Feige JN. Aging Disrupts Muscle Stem Cell Function by Impairing Matricellular WISP1 Secretion from Fibro-Adipogenic Progenitors. Cell Stem Cell 2019; 24:433-446.e7. [PMID: 30686765 DOI: 10.1016/j.stem.2018.12.014] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 10/15/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022]
Abstract
Research on age-related regenerative failure of skeletal muscle has extensively focused on the phenotypes of muscle stem cells (MuSCs). In contrast, the impact of aging on regulatory cells in the MuSC niche remains largely unexplored. Here, we demonstrate that aging impairs the function of mouse fibro-adipogenic progenitors (FAPs) and thereby indirectly affects the myogenic potential of MuSCs. Using transcriptomic profiling, we identify WNT1 Inducible Signaling Pathway Protein 1 (WISP1) as a FAP-derived matricellular signal that is lost during aging. WISP1 is required for efficient muscle regeneration and controls the expansion and asymmetric commitment of MuSCs through Akt signaling. Transplantation of young FAPs or systemic treatment with WISP1 restores the myogenic capacity of MuSCs in aged mice and rescues skeletal muscle regeneration. Our work establishes that loss of WISP1 from FAPs contributes to MuSC dysfunction in aged skeletal muscles and demonstrates that this mechanism can be targeted to rejuvenate myogenesis.
Collapse
Affiliation(s)
- Laura Lukjanenko
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sonia Karaz
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Pascal Stuelsatz
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Uxia Gurriaran-Rodriguez
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Joris Michaud
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Gabriele Dammone
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Federico Sizzano
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Omid Mashinchian
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sara Ancel
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | | | - Sophie Liot
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, Lyon, France
| | - Guillaume Jacot
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | | | - Frederic Raymond
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | | | - Alessio Palini
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Benedicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, Lyon, France
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - C Florian Bentzinger
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jerome N Feige
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
169
|
Roh DS, Panayi AC, Bhasin S, Orgill DP, Sinha I. Implications of Aging in Plastic Surgery. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2019; 7:e2085. [PMID: 30859042 PMCID: PMC6382222 DOI: 10.1097/gox.0000000000002085] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022]
Abstract
Given the rapidly aging population, investigating the effect of age on plastic surgery outcomes is imperative. Despite this, the topic has received relatively little attention. Furthermore, there appears to be little integration between the basic scientists investigating the mechanisms of aging and the plastic surgeons providing the majority of "antiaging" therapies. This review first provides a description of the effects and mechanisms of aging in 5 types of tissue: skin, adipose tissue, muscles, bones and tendons, and nervous tissue followed by an overview of the basic mechanisms underlying aging, presenting the currently proposed cellular and molecular theories. Finally, the impact of aging, as well as frailty, on plastic surgery outcomes is explored by focusing on 5 different topics: general wound healing and repair of cutaneous tissue, reconstruction of soft tissue, healing of bones and tendons, healing of peripheral nerves, and microsurgical reconstruction. We find mixed reports on the effect of aging or frailty on outcomes in plastic surgery, which we hypothesize to be due to exclusion of aged and frail patients from surgery as well as due to outcomes that reported no postsurgical issues with aged patients. As plastic surgeons continue to interact more with the growing elderly population, a better appreciation of the underlying mechanisms and outcomes related to aging and a clear distinction between chronological age and frailty can promote better selection of patients, offering appropriate patients surgery to improve an aged appearance, and declining interventions in inappropriate patients.
Collapse
Affiliation(s)
- Danny S Roh
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Shalender Bhasin
- Division of Endocrinology, Brigham and Women's Hospital, Boston, Mass
| | - Dennis P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Mass
| | - Indranil Sinha
- Division of Plastic Surgery, Brigham and Women's Hospital, Boston, Mass
| |
Collapse
|
170
|
Tseng C, Sinha K, Pan H, Cui Y, Guo P, Lin CY, Yang F, Deng Z, Eltzschig HK, Lu A, Huard J. Markers of Accelerated Skeletal Muscle Regenerative Response in Murphy Roths Large Mice: Characteristics of Muscle Progenitor Cells and Circulating Factors. Stem Cells 2019; 37:357-367. [PMID: 30537304 DOI: 10.1002/stem.2957] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/06/2018] [Accepted: 11/14/2018] [Indexed: 12/19/2022]
Abstract
The "super-healing" Murphy Roths Large (MRL/MpJ) mouse possesses a superior regenerative capacity for repair of many tissues, which makes it an excellent animal model for studying molecular and cellular mechanisms during tissue regeneration. As the role of muscle progenitor cells (MPCs) in muscle-healing capacity of MRL/MpJ mice has not been previously studied, we investigated the muscle regenerative capacity of MRL/MpJ mice following muscle injury, and the results were compared to results from C57BL/6J (B6) age-matched control mice. Our results show that muscle healing upon cardiotoxin injury was accelerated in MRL/MpJ mice and characterized by reduced necrotic muscle area, reduced macrophage infiltration, and more regenerated myofibers (embryonic myosin heavy chain+/centronucleated fibers) at 3, 5, and 12 days postinjury, when compared to B6 age-matched control mice. These observations were associated with enhanced function of MPCs, including improved cell proliferation, differentiation, and resistance to stress, as well as increased muscle regenerative potential when compared to B6 MPCs. Mass spectrometry of serum proteins revealed higher levels of circulating antioxidants in MRL/MpJ mice when compared to B6 mice. Indeed, we found relatively higher gene expression of superoxide dismutase 1 (Sod1) and catalase (Cat) in MRL/MpJ MPCs. Depletion of Sod1 or Cat by small interfering RNA impaired myogenic potential of MRL/MpJ MPCs, indicating a role for these antioxidants in muscle repair. Taken together, these findings provide evidence that improved function of MPCs and higher levels of circulating antioxidants play important roles in accelerating muscle-healing capacity of MRL/MpJ mice. Stem Cells 2019;37:357-367.
Collapse
Affiliation(s)
- Chieh Tseng
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Krishna Sinha
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Haiying Pan
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Yan Cui
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ping Guo
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Chih Yi Lin
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Fan Yang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Zhenhan Deng
- Department of Sports Medicine, Shenzhen Second People's Hospital, Shenzhen, Guangzhou, People's Republic of China
| | - Holger K Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Aiping Lu
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| |
Collapse
|
171
|
Perdiguero E, Moiseeva V, Muñoz-Cánoves P. Simultaneous Isolation of Stem and Niche Cells of Skeletal Muscle: Applicability for Aging Studies. Methods Mol Biol 2019; 2045:13-23. [PMID: 30771188 DOI: 10.1007/7651_2019_210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The maintenance of adult stem cells in their normal quiescent state depends on intrinsic factors and extrinsic signals originating from their microenvironment (also known as the stem cell niche). In skeletal muscle, its stem cells (satellite cells) lose their regenerative potential with aging, and this has been attributed, at least in part, to both age-associated changes in the satellite cells as in the niche cells, which include resident fibro-adipogenic progenitors (FAPs), macrophages, and endothelial cells, among others. To understand the regenerative decline of skeletal muscle with aging, there is a need for methods to specifically isolate stem and niche cells from resting muscle. Here we describe a fluorescence-activated cell sorting (FACS) protocol to simultaneously isolate discrete populations of satellite cells and niche cells from skeletal muscle of aging mice.
Collapse
Affiliation(s)
- Eusebio Perdiguero
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.
| | - Victoria Moiseeva
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Pura Muñoz-Cánoves
- Cell Biology Group, Department of Experimental and Health Sciences, Pompeu Fabra University (UPF), CIBER on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain.
- Spanish National Center on Cardiovascular Research (CNIC), Madrid, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
172
|
Franco I, Fernandez-Gonzalo R, Vrtačnik P, Lundberg TR, Eriksson M, Gustafsson T. Healthy skeletal muscle aging: The role of satellite cells, somatic mutations and exercise. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 346:157-200. [DOI: 10.1016/bs.ircmb.2019.03.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
173
|
Epigenetic Erosion in Adult Stem Cells: Drivers and Passengers of Aging. Cells 2018; 7:cells7120237. [PMID: 30501028 PMCID: PMC6316114 DOI: 10.3390/cells7120237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023] Open
Abstract
In complex organisms, stem cells are key for tissue maintenance and regeneration. Adult stem cells replenish continuously dividing tissues of the epithelial and connective types, whereas in non-growing muscle and nervous tissues, they are mainly activated upon injury or stress. In addition to replacing deteriorated cells, adult stem cells have to prevent their exhaustion by self-renewal. There is mounting evidence that both differentiation and self-renewal are impaired upon aging, leading to tissue degeneration and functional decline. Understanding the molecular pathways that become deregulate in old stem cells is crucial to counteract aging-associated tissue impairment. In this review, we focus on the epigenetic mechanisms governing the transition between quiescent and active states, as well as the decision between self-renewal and differentiation in three different stem cell types, i.e., spermatogonial stem cells, hematopoietic stem cells, and muscle stem cells. We discuss the epigenetic events that channel stem cell fate decisions, how this epigenetic regulation is altered with age, and how this can lead to tissue dysfunction and disease. Finally, we provide short prospects of strategies to preserve stem cell function and thus promote healthy aging.
Collapse
|
174
|
Oncogenic Amplification of Zygotic Dux Factors in Regenerating p53-Deficient Muscle Stem Cells Defines a Molecular Cancer Subtype. Cell Stem Cell 2018; 23:794-805.e4. [PMID: 30449715 DOI: 10.1016/j.stem.2018.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/27/2018] [Accepted: 10/08/2018] [Indexed: 01/09/2023]
Abstract
The identity of tumor-initiating cells in many cancer types is unknown. Tumors often express genes associated with embryonic development, although the contributions of zygotic programs to tumor initiation and formation are poorly understood. Here, we show that regeneration-induced loss of quiescence in p53-deficient muscle stem cells (MuSCs) results in rhabdomyosarcoma formation with 100% penetrance. Genomic analyses of purified tumor cells revealed spontaneous and discrete oncogenic amplifications in MuSCs that drive tumorigenesis, including, but not limited to, the amplification of the cleavage-stage Dux transcription factor (TF) Duxbl. We further found that Dux factors drive an early embryonic gene signature that defines a molecular subtype across a broad range of human cancers. Duxbl initiates tumorigenesis by enforcing a mesenchymal-to-epithelial transition, and targeted inactivation of Duxbl specifically in Duxbl-expressing tumor cells abolishes their expansion. These findings reveal how regeneration and genomic instability can interact to activate zygotic genes that drive tumor initiation and growth.
Collapse
|
175
|
Cho CH, Lee KJ, Lee EH. With the greatest care, stromal interaction molecule (STIM) proteins verify what skeletal muscle is doing. BMB Rep 2018; 51:378-387. [PMID: 29898810 PMCID: PMC6130827 DOI: 10.5483/bmbrep.2018.51.8.128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Indexed: 12/11/2022] Open
Abstract
Skeletal muscle contracts or relaxes to maintain the body position and locomotion. For the contraction and relaxation of skeletal muscle, Ca2+ in the cytosol of skeletal muscle fibers acts as a switch to turn on and off a series of contractile proteins. The cytosolic Ca2+ level in skeletal muscle fibers is governed mainly by movements of Ca2+ between the cytosol and the sarcoplasmic reticulum (SR). Store-operated Ca2+ entry (SOCE), a Ca2+ entryway from the extracellular space to the cytosol, has gained a significant amount of attention from muscle physiologists. Orai1 and stromal interaction molecule 1 (STIM1) are the main protein identities of SOCE. This mini-review focuses on the roles of STIM proteins and SOCE in the physiological and pathophysiological functions of skeletal muscle and in their correlations with recently identified proteins, as well as historical proteins that are known to mediate skeletal muscle function.
Collapse
Affiliation(s)
- Chung-Hyun Cho
- Department of Pharmacology, College of Medicine, Seoul National University, Seoul 08826, Korea
| | - Keon Jin Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea; Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea; Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
176
|
Dorsal Root Ganglion Maintains Stemness of Bone Marrow Mesenchymal Stem Cells by Enhancing Autophagy through the AMPK/mTOR Pathway in a Coculture System. Stem Cells Int 2018; 2018:8478953. [PMID: 30363977 PMCID: PMC6186314 DOI: 10.1155/2018/8478953] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/10/2018] [Accepted: 08/14/2018] [Indexed: 12/28/2022] Open
Abstract
Our previous studies found that sensory nerve tracts implanted in tissue-engineered bone (TEB) could result in better osteogenesis. To explore the mechanism of the sensory nerve promoting osteogenesis in TEB in vitro, a transwell coculture experiment was designed between dorsal root ganglion (DRG) cells and bone marrow mesenchymal stem cells (BMSCs). BMSC proliferation was determined by CCK8 assay, and osteo-, chondro-, and adipogenic differentiation were assessed by alizarin red, alcian blue, and oil red staining. We found that the proliferation and multipotent differentiation of BMSCs were all enhanced in the coculture group compared to the BMSCs group. Crystal violet staining showed that the clone-forming ability of BMSCs in the coculture group was also enhanced and mRNA levels of Sox2, Nanog, and Oct4 were significantly upregulated in the coculture group. Moreover, the autophagy level of BMSCs, regulating their stemness, was promoted in the coculture group, mediated by the AMPK/mTOR pathway. In addition, AMPK inhibitor compound C could significantly downregulate the protein expression of LC3 and the mRNA level of stemness genes in the coculture group. Finally, we found that the NK1 receptor antagonist, aprepitant, could partly block this effect, which indicated that substance P played an important role in the effect. Together, we conclude that DRG could maintain the stemness of BMSCs by enhancing autophagy through the AMPK/mTOR pathway in a transwell coculture system, which may help explain the better osteogenesis after implantation of the sensory nerve into TEB.
Collapse
|
177
|
Maleiner B, Tomasch J, Heher P, Spadiut O, Rünzler D, Fuchs C. The Importance of Biophysical and Biochemical Stimuli in Dynamic Skeletal Muscle Models. Front Physiol 2018; 9:1130. [PMID: 30246791 PMCID: PMC6113794 DOI: 10.3389/fphys.2018.01130] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/30/2018] [Indexed: 12/31/2022] Open
Abstract
Classical approaches to engineer skeletal muscle tissue based on current regenerative and surgical procedures still do not meet the desired outcome for patient applications. Besides the evident need to create functional skeletal muscle tissue for the repair of volumetric muscle defects, there is also growing demand for platforms to study muscle-related diseases, such as muscular dystrophies or sarcopenia. Currently, numerous studies exist that have employed a variety of biomaterials, cell types and strategies for maturation of skeletal muscle tissue in 2D and 3D environments. However, researchers are just at the beginning of understanding the impact of different culture settings and their biochemical (growth factors and chemical changes) and biophysical cues (mechanical properties) on myogenesis. With this review we intend to emphasize the need for new in vitro skeletal muscle (disease) models to better recapitulate important structural and functional aspects of muscle development. We highlight the importance of choosing appropriate system components, e.g., cell and biomaterial type, structural and mechanical matrix properties or culture format, and how understanding their interplay will enable researchers to create optimized platforms to investigate myogenesis in healthy and diseased tissue. Thus, we aim to deliver guidelines for experimental designs to allow estimation of the potential influence of the selected skeletal muscle tissue engineering setup on the myogenic outcome prior to their implementation. Moreover, we offer a workflow to facilitate identifying and selecting different analytical tools to demonstrate the successful creation of functional skeletal muscle tissue. Ultimately, a refinement of existing strategies will lead to further progression in understanding important aspects of muscle diseases, muscle aging and muscle regeneration to improve quality of life of patients and enable the establishment of new treatment options.
Collapse
Affiliation(s)
- Babette Maleiner
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Janine Tomasch
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Philipp Heher
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology/AUVA Research Center, Vienna, Austria.,Trauma Care Consult GmbH, Vienna, Austria
| | - Oliver Spadiut
- Institute of Chemical Engineering, Vienna University of Technology, Vienna, Austria
| | - Dominik Rünzler
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christiane Fuchs
- Department of Biochemical Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria.,The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
178
|
Han WM, Anderson SE, Mohiuddin M, Barros D, Nakhai SA, Shin E, Amaral IF, Pêgo AP, García AJ, Jang YC. Synthetic matrix enhances transplanted satellite cell engraftment in dystrophic and aged skeletal muscle with comorbid trauma. SCIENCE ADVANCES 2018; 4:eaar4008. [PMID: 30116776 PMCID: PMC6093653 DOI: 10.1126/sciadv.aar4008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 07/11/2018] [Indexed: 05/29/2023]
Abstract
Muscle satellite cells (MuSCs) play a central role in muscle regeneration, but their quantity and function decline with comorbidity of trauma, aging, and muscle diseases. Although transplantation of MuSCs in traumatically injured muscle in the comorbid context of aging or pathology is a strategy to boost muscle regeneration, an effective cell delivery strategy in these contexts has not been developed. We engineered a synthetic hydrogel-based matrix with optimal mechanical, cell-adhesive, and protease-degradable properties that promotes MuSC survival, proliferation, and differentiation. Furthermore, we establish a biomaterial-mediated cell delivery strategy for treating muscle trauma, where intramuscular injections may not be applicable. Delivery of MuSCs in the engineered matrix significantly improved in vivo cell survival, proliferation, and engraftment in nonirradiated and immunocompetent muscles of aged and dystrophic mice compared to collagen gels and cell-only controls. This platform may be suitable for treating craniofacial and limb muscle trauma, as well as postoperative wounds of elderly and dystrophic patients.
Collapse
Affiliation(s)
- Woojin M. Han
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Shannon E. Anderson
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mahir Mohiuddin
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Daniela Barros
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Shadi A. Nakhai
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Eunjung Shin
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Isabel Freitas Amaral
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Ana Paula Pêgo
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Andrés J. García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Young C. Jang
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
179
|
Maintaining bovine satellite cells stemness through p38 pathway. Sci Rep 2018; 8:10808. [PMID: 30018348 PMCID: PMC6050236 DOI: 10.1038/s41598-018-28746-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/26/2018] [Indexed: 12/01/2022] Open
Abstract
Isolating and maintaining the appropriate stem cell for large scale cell culture is essential in tissue engineering or food production. For bovine satellite cells an optimized isolation and purification protocol is lacking and there is also no detailed understanding on the factors that maintain stemness of these cells. Here, we set up a fluorescence-activated cell sorting strategy to enrich bovine satellite cells. We found that p38-MAPK signalling is activated and PAX7 expression is gradually lost during satellite cell proliferation. The p38 inhibitor (SB203580) treatment maintained PAX7 expression but inhibited the fusion of satellite cells in a concentration-dependent way in short-term incubation. The mechanism of p38 inhibition was confirmed by inhibiting canonical p38 signalling, i.e. HSP27. Long-term culture with an appropriate concentration of p38i enhanced the proliferation and PAX7 expression, while the differentiation capacity recovered and was enhanced compared to vehicle control. These studies indicate that bovine satellite cells maintenance depends on cell purity and p38 MAPK signalling. Inhibition of p38 MAPK signaling is a promising strategy to facilitate large scale cell expansion of primary cells for tissue engineering and cultured meat purposes.
Collapse
|
180
|
Wosczyna MN, Rando TA. A Muscle Stem Cell Support Group: Coordinated Cellular Responses in Muscle Regeneration. Dev Cell 2018; 46:135-143. [PMID: 30016618 PMCID: PMC6075730 DOI: 10.1016/j.devcel.2018.06.018] [Citation(s) in RCA: 202] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 06/12/2018] [Accepted: 06/20/2018] [Indexed: 01/11/2023]
Abstract
Skeletal muscle has an extraordinary regenerative capacity due to the activity of tissue-specific muscle stem cells. Consequently, these cells have received the most attention in studies investigating the cellular processes of skeletal muscle regeneration. However, efficient capacity to rebuild this tissue also depends on additional cells in the local milieu, as disrupting their normal contributions often leads to incomplete regeneration. Here, we review these additional cells that contribute to the regenerative process. Understanding the complex interactions between and among these cell populations has the potential to lead to therapies that will help promote normal skeletal muscle regeneration under conditions in which this process is suboptimal.
Collapse
Affiliation(s)
- Michael N Wosczyna
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| |
Collapse
|
181
|
BGP-15 improves contractile function of regenerating soleus muscle. J Muscle Res Cell Motil 2018; 39:25-34. [PMID: 29948663 DOI: 10.1007/s10974-018-9495-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/05/2018] [Indexed: 02/04/2023]
Abstract
This study investigated the effect of the heat shock protein inducer O-[3-piperidino-2-hydroxy-1-propyl]-nicotinic amidoxime (BGP-15) on the morphology and contractile function of regenerating soleus muscles from mice. Cryolesioned soleus muscles from young mice treated daily with BGP-15 (15 mg/Kg) were evaluated on post-cryolesion day 10. At this time point, there was a significant decrease in the cross-sectional area of regenerating myofibers, maximal force, specific tetanic force, and fatigue resistance of regenerating soleus muscles. BGP-15 did not reverse the decrease in myofiber cross-sectional area but effectively prevented the reduction in tetanic force and fatigue resistance of regenerating muscles. In addition, BGP-15 treatment increased the expression of embryonic myosin heavy chain (e-MyHC), MyHC-II and MyHC-I in regenerating muscles. Although BGP-15 did not alter voltage dependent anion-selective channel 2 (VDAC2) expression in cryolesioned muscles, it was able to increase inducible 70-kDa heat shock protein (HSP70) expression. Our results suggest that BGP-15 improves strength recovery in regenerating soleus muscles by accelerating the re-expression of adult MyHC-II and MyHC-I isoforms and HSP70 induction. The beneficial effects of BGP-15 on the contractile function of regenerating muscles reinforce the potential of this molecule to be used as a therapeutic agent.
Collapse
|
182
|
A self-made quiescent niche. Nat Rev Mol Cell Biol 2018; 19:416-417. [PMID: 29875372 DOI: 10.1038/s41580-018-0031-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
183
|
Dinulovic I, Furrer R, Handschin C. Plasticity of the Muscle Stem Cell Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:141-169. [PMID: 29204832 DOI: 10.1007/978-3-319-69194-7_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Satellite cells (SCs) are adult muscle stem cells capable of repairing damaged and creating new muscle tissue throughout life. Their functionality is tightly controlled by a microenvironment composed of a wide variety of factors, such as numerous secreted molecules and different cell types, including blood vessels, oxygen, hormones, motor neurons, immune cells, cytokines, fibroblasts, growth factors, myofibers, myofiber metabolism, the extracellular matrix and tissue stiffness. This complex niche controls SC biology-quiescence, activation, proliferation, differentiation or renewal and return to quiescence. In this review, we attempt to give a brief overview of the most important players in the niche and their mutual interaction with SCs. We address the importance of the niche to SC behavior under physiological and pathological conditions, and finally survey the significance of an artificial niche both for basic and translational research purposes.
Collapse
|
184
|
Kim YA, Jin SW, Oh SH, Lee GH, Pham HT, Choi JH, Chung YC, Lee WL, Kim SK, Jeong HG. Platycodon grandiflorum-derived saponin enhances exercise function, skeletal muscle protein synthesis, and mitochondrial function. Food Chem Toxicol 2018; 118:94-104. [PMID: 29723585 DOI: 10.1016/j.fct.2018.04.062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/25/2018] [Accepted: 04/28/2018] [Indexed: 10/17/2022]
Abstract
Lower physical performance is an important risk factor in hypokinetic-related chronic disease, metabolic syndrome, and muscle atrophy. Our previous research demonstrated that Platycodon grandiflorum-derived saponin (PS) protects against eccentric exercise-induced muscle damage and mitochondrial function-related peroxisomal acyl-coenzme A oxidase (ACOX-1) and carnitine palmitoyltransferase (CPT-1) in high-fat diet-induced non-alcoholic steatohepatitis, and it inhibits osteoclast differentiation. However, the effects of PS on physical performance remain unknown. Therefore, we investigated whether PS enhances physical activity and skeletal muscle function. Supplementation with PS (2 mg/kg for 4 weeks) increased grip strength, wheel running repetition, and time to exhaustion in treadmill and swimming exercises. Marked increases in the synthesis of skeletal muscle proteins and muscle stem cell-related paired-box 7 (PAX7) were observed, and a decrease in the negative regulator myostatin was associated with enhanced muscle regeneration. Furthermore, PS induced expression of mitochondrial function proteins, including OXPHOS-III and -IV, in vivo and in vitro. These results suggest that PS enhances exercise function by ameliorating skeletal muscle protein synthesis and mitochondrial function.
Collapse
Affiliation(s)
- Yong An Kim
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Sun Woo Jin
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Suck Hoon Oh
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Gi Ho Lee
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Hoa Thi Pham
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jae Ho Choi
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Young Chul Chung
- Department of Food Science, International University of Korea, Jinju, Republic of Korea
| | - Wang Lok Lee
- Department of Sport Science, Chungnam National University, Daejeon, Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Hye Gwang Jeong
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
185
|
Wang YN, Yang WC, Li PW, Wang HB, Zhang YY, Zan LS. Myocyte enhancer factor 2A promotes proliferation and its inhibition attenuates myogenic differentiation via myozenin 2 in bovine skeletal muscle myoblast. PLoS One 2018; 13:e0196255. [PMID: 29698438 PMCID: PMC5919640 DOI: 10.1371/journal.pone.0196255] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 04/09/2018] [Indexed: 12/18/2022] Open
Abstract
Myocyte enhancer factor 2A (MEF2A) is widely distributed in various tissues or organs and plays crucial roles in multiple biological processes. To examine the potential effects of MEF2A on skeletal muscle myoblast, the functional role of MFE2A in myoblast proliferation and differentiation was investigated. In this study, we found that the mRNA expression level of Mef2a was dramatically increased during the myogenesis of bovine skeletal muscle primary myoblast. Overexpression of MEF2A significantly promoted myoblast proliferation, while knockdown of MEF2A inhibited the proliferation and differentiation of myoblast. RT-PCR and western blot analysis revealed that this positive effect of MEF2A on the proliferation of myoblast was carried out by triggering cell cycle progression by activating CDK2 protein expression. Besides, MEF2A was found to be an important transcription factor that bound to the myozenin 2 (MyoZ2) proximal promoter and performed upstream of MyoZ2 during myoblast differentiation. This study provides the first experimental evidence that MEF2A is a positive regulator in skeletal muscle myoblast proliferation and suggests that MEF2A regulates myoblast differentiation via regulating MyoZ2.
Collapse
Affiliation(s)
- Ya-Ning Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- National Beef Cattle Improvement Center in China, Yangling, Shaanxi, P. R. China
| | - Wu-Cai Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- National Beef Cattle Improvement Center in China, Yangling, Shaanxi, P. R. China
| | - Pei-Wei Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- National Beef Cattle Improvement Center in China, Yangling, Shaanxi, P. R. China
| | - Hong-Bao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- National Beef Cattle Improvement Center in China, Yangling, Shaanxi, P. R. China
| | - Ying-Ying Zhang
- Animal Husbandry and Veterinary Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, P. R. China
| | - Lin-Sen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- National Beef Cattle Improvement Center in China, Yangling, Shaanxi, P. R. China
- National and Provincial Joint Engineering Research Center of Modern Cattle Biotechnology and Applications, Yangling, Shaanxi, P. R. China
- * E-mail:
| |
Collapse
|
186
|
Thomas JD, Oliveira R, Sznajder ŁJ, Swanson MS. Myotonic Dystrophy and Developmental Regulation of RNA Processing. Compr Physiol 2018; 8:509-553. [PMID: 29687899 PMCID: PMC11323716 DOI: 10.1002/cphy.c170002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Myotonic dystrophy (DM) is a multisystemic disorder caused by microsatellite expansion mutations in two unrelated genes leading to similar, yet distinct, diseases. DM disease presentation is highly variable and distinguished by differences in age-of-onset and symptom severity. In the most severe form, DM presents with congenital onset and profound developmental defects. At the molecular level, DM pathogenesis is characterized by a toxic RNA gain-of-function mechanism that involves the transcription of noncoding microsatellite expansions. These mutant RNAs disrupt key cellular pathways, including RNA processing, localization, and translation. In DM, these toxic RNA effects are predominantly mediated through the modulation of the muscleblind-like and CUGBP and ETR-3-like factor families of RNA binding proteins (RBPs). Dysfunction of these RBPs results in widespread RNA processing defects culminating in the expression of developmentally inappropriate protein isoforms in adult tissues. The tissue that is the focus of this review, skeletal muscle, is particularly sensitive to mutant RNA-responsive perturbations, as patients display a variety of developmental, structural, and functional defects in muscle. Here, we provide a comprehensive overview of DM1 and DM2 clinical presentation and pathology as well as the underlying cellular and molecular defects associated with DM disease onset and progression. Additionally, fundamental aspects of skeletal muscle development altered in DM are highlighted together with ongoing and potential therapeutic avenues to treat this muscular dystrophy. © 2018 American Physiological Society. Compr Physiol 8:509-553, 2018.
Collapse
Affiliation(s)
- James D. Thomas
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Ruan Oliveira
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Łukasz J. Sznajder
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| | - Maurice S. Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
187
|
Ling YH, Sui MH, Zheng Q, Wang KY, Wu H, Li WY, Liu Y, Chu MX, Fang FG, Xu LN. miR-27b regulates myogenic proliferation and differentiation by targeting Pax3 in goat. Sci Rep 2018; 8:3909. [PMID: 29500394 PMCID: PMC5834623 DOI: 10.1038/s41598-018-22262-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/19/2018] [Indexed: 02/08/2023] Open
Abstract
This study found that miR-27 is expressed in muscle and regulates muscle proliferation and differentiation. We explored the function and regulatory mechanism of miR-27b in goat muscle proliferation and differentiation. Compared with the Boer goat, higher expression of miR-27b was observed in all of the collected muscle tissues of Anhuai goat, excluding the kidney, whereas the opposite expression pattern was observed for Pax3, which showed lower expression in Anhuai goat. Expression of miR-27b decreased gradually during the proliferation of skeletal muscle satellite cells in Anhuai goat and increased during differentiation; however, the expression pattern of Pax3 was opposite. The regulatory activity of miR-27b demonstrated that miR-27b inhibited the proliferation of skeletal muscle satellite cells, but promoted their differentiation. Moreover, function research demonstrated that Pax3 negatively regulated myogenic differentiation of goat skeletal muscle satellite cells, but accelerated their proliferation. The results of a dual-luciferase reporter analysis showed that miR-27b directly targeted the 3’-untranslated regions of Pax3 mRNA, and western blot and immunofluorescence staining analyses showed that miR-27b inhibited expression of the Pax3 protein. In goats, miR-27b can regulate myogenic proliferation and differentiation by targeting Pax3.
Collapse
Affiliation(s)
- Ying-Hui Ling
- College of Animal Science and Technology, Anhui Agricultural University, Anhui Hefei, China.,Local animal genetic resources conservation and biobreeding laboratory of Anhui province, Anhui Hefei, China
| | - Meng-Hua Sui
- College of Animal Science and Technology, Anhui Agricultural University, Anhui Hefei, China.,Local animal genetic resources conservation and biobreeding laboratory of Anhui province, Anhui Hefei, China
| | - Qi Zheng
- College of Animal Science and Technology, Anhui Agricultural University, Anhui Hefei, China.,Local animal genetic resources conservation and biobreeding laboratory of Anhui province, Anhui Hefei, China
| | - Kang-Yan Wang
- College of Animal Science and Technology, Anhui Agricultural University, Anhui Hefei, China.,Local animal genetic resources conservation and biobreeding laboratory of Anhui province, Anhui Hefei, China
| | - Hao Wu
- College of Animal Science and Technology, Anhui Agricultural University, Anhui Hefei, China.,Local animal genetic resources conservation and biobreeding laboratory of Anhui province, Anhui Hefei, China
| | - Wen-Yong Li
- Key Laboratory of Embryo Development and Reproductive Regulation of Anhui Province, Fuyang Normal University, Fuyang, Anhui, 236037, China
| | - Yong Liu
- Key Laboratory of Embryo Development and Reproductive Regulation of Anhui Province, Fuyang Normal University, Fuyang, Anhui, 236037, China
| | - Ming-Xing Chu
- Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, CAAS, Beijing, 100193, China
| | - Fu-Gui Fang
- College of Animal Science and Technology, Anhui Agricultural University, Anhui Hefei, China.,Local animal genetic resources conservation and biobreeding laboratory of Anhui province, Anhui Hefei, China
| | - Li-Na Xu
- College of Animal Science and Technology, Anhui Agricultural University, Anhui Hefei, China. .,Institute of Plant Protection and Agro-Products Safety, Anhui Academy of Agricultural Sciences, Hefei, Anhui, 230031, China.
| |
Collapse
|
188
|
Franco I, Johansson A, Olsson K, Vrtačnik P, Lundin P, Helgadottir HT, Larsson M, Revêchon G, Bosia C, Pagnani A, Provero P, Gustafsson T, Fischer H, Eriksson M. Somatic mutagenesis in satellite cells associates with human skeletal muscle aging. Nat Commun 2018; 9:800. [PMID: 29476074 PMCID: PMC5824957 DOI: 10.1038/s41467-018-03244-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/26/2018] [Indexed: 01/06/2023] Open
Abstract
Human aging is associated with a decline in skeletal muscle (SkM) function and a reduction in the number and activity of satellite cells (SCs), the resident stem cells. To study the connection between SC aging and muscle impairment, we analyze the whole genome of single SC clones of the leg muscle vastus lateralis from healthy individuals of different ages (21–78 years). We find an accumulation rate of 13 somatic mutations per genome per year, consistent with proliferation of SCs in the healthy adult muscle. SkM-expressed genes are protected from mutations, but aging results in an increase in mutations in exons and promoters, targeting genes involved in SC activity and muscle function. In agreement with SC mutations affecting the whole tissue, we detect a missense mutation in a SC propagating to the muscle. Our results suggest somatic mutagenesis in SCs as a driving force in the age-related decline of SkM function. Aging skeletal muscle shows declining numbers and activity of satellite cells. Here, Franco et al. show that in satellite cells of the human leg muscle vastus lateralis, somatic mutations accumulate with age and that these mutations become enriched in exons and promoters of genes involved in muscle function.
Collapse
Affiliation(s)
- Irene Franco
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, 14157, Huddinge, Sweden.
| | - Anna Johansson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, 75237, Uppsala, Sweden
| | - Karl Olsson
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, 14186, Huddinge, Sweden
| | - Peter Vrtačnik
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, 14157, Huddinge, Sweden
| | - Pär Lundin
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, 14157, Huddinge, Sweden.,Science for Life Laboratory, Department of Biochemistry and Biophysics (DBB), Stockholm University, 10691, Stockholm, Sweden
| | - Hafdis T Helgadottir
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, 14157, Huddinge, Sweden
| | - Malin Larsson
- Science for Life Laboratory, Department of Physics, Chemistry and Biology, Linköping University, 58183, Linköping, Sweden
| | - Gwladys Revêchon
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, 14157, Huddinge, Sweden
| | - Carla Bosia
- Italian Institute for Genomic Medicine (IIGM), 10126, Turin, Italy.,Department of Applied Science and Technology, Politecnico di Torino, 10129, Turin, Italy
| | - Andrea Pagnani
- Italian Institute for Genomic Medicine (IIGM), 10126, Turin, Italy.,Department of Applied Science and Technology, Politecnico di Torino, 10129, Turin, Italy
| | - Paolo Provero
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, 10126, Turin, Italy.,Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Thomas Gustafsson
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, 14186, Huddinge, Sweden
| | - Helene Fischer
- Division of Clinical Physiology, Department of Laboratory Medicine, Karolinska Institutet, 14186, Huddinge, Sweden
| | - Maria Eriksson
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, 14157, Huddinge, Sweden.
| |
Collapse
|
189
|
Bernacchioni C, Cencetti F, Ouro A, Bruno M, Gomez-Muñoz A, Donati C, Bruni P. Lysophosphatidic Acid Signaling Axis Mediates Ceramide 1-Phosphate-Induced Proliferation of C2C12 Myoblasts. Int J Mol Sci 2018; 19:ijms19010139. [PMID: 29300303 PMCID: PMC5796088 DOI: 10.3390/ijms19010139] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 12/23/2017] [Accepted: 12/28/2017] [Indexed: 02/07/2023] Open
Abstract
Sphingolipids are not only crucial for membrane architecture but act as critical regulators of cell functions. The bioactive sphingolipid ceramide 1-phosphate (C1P), generated by the action of ceramide kinase, has been reported to stimulate cell proliferation, cell migration and to regulate inflammatory responses via activation of different signaling pathways. We have previously shown that skeletal muscle is a tissue target for C1P since the phosphosphingolipid plays a positive role in myoblast proliferation implying a role in muscle regeneration. Skeletal muscle displays strong capacity of regeneration thanks to the presence of quiescent adult stem cells called satellite cells that upon trauma enter into the cell cycle and start proliferating. However, at present, the exact molecular mechanism by which C1P triggers its mitogenic effect in myoblasts is lacking. Here, we report for the first time that C1P stimulates C2C12 myoblast proliferation via lysophosphatidic acid (LPA) signaling axis. Indeed, C1P subsequently to phospholipase A2 activation leads to LPA1 and LPA3 engagement, which in turn drive Akt (protein kinase B) and ERK1/2 (extracellular signal-regulated kinases 1/2) activation, thus stimulating DNA synthesis. The present findings shed new light on the key role of bioactive sphingolipids in skeletal muscle and provide further support to the notion that these pleiotropic molecules might be useful therapeutic targets for skeletal muscle regeneration.
Collapse
Affiliation(s)
- Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| | - Alberto Ouro
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48080 Bilbao, Spain.
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Marina Bruno
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
| | - Antonio Gomez-Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48080 Bilbao, Spain.
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale GB Morgagni 50, 50134 Firenze, Italy.
- Istituto Interuniversitario di Miologia (IIM), Italy.
| |
Collapse
|
190
|
Muscle Atrophy: Present and Future. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:605-624. [DOI: 10.1007/978-981-13-1435-3_29] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
191
|
Past, Present, and Future Perspective of Targeting Myostatin and Related Signaling Pathways to Counteract Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:153-206. [DOI: 10.1007/978-981-13-1435-3_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
192
|
Rahmati M, Pennisi CP, Mobasheri A, Mozafari M. Bioengineered Scaffolds for Stem Cell Applications in Tissue Engineering and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1107:73-89. [DOI: 10.1007/5584_2018_215] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
193
|
Randrianarison-Huetz V, Papaefthymiou A, Herledan G, Noviello C, Faradova U, Collard L, Pincini A, Schol E, Decaux JF, Maire P, Vassilopoulos S, Sotiropoulos A. Srf controls satellite cell fusion through the maintenance of actin architecture. J Cell Biol 2017; 217:685-700. [PMID: 29269426 PMCID: PMC5800804 DOI: 10.1083/jcb.201705130] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 10/20/2017] [Accepted: 11/21/2017] [Indexed: 01/17/2023] Open
Abstract
This work describes a crucial role for the transcription factor Srf and F-actin scaffold to drive muscle stem cell fusion in vitro and in vivo and provides evidence of how actin cytoskeleton architecture affects myoblast fusion in vertebrates. Satellite cells (SCs) are adult muscle stem cells that are mobilized when muscle homeostasis is perturbed. Here, we show that serum response factor (Srf) is needed for optimal SC-mediated hypertrophic growth. We identified Srf as a master regulator of SC fusion required in both fusion partners, whereas it was dispensable for SC proliferation and differentiation. We show that SC-specific Srf deletion leads to impaired actin cytoskeleton and report the existence of finger-like actin–based protrusions at fusion sites in vertebrates that were notoriously absent in fusion-defective myoblasts lacking Srf. Restoration of a polymerized actin network by overexpression of an α-actin isoform in Srf mutant SCs rescued their fusion with a control cell in vitro and in vivo and reestablished overload-induced muscle growth. These findings demonstrate the importance of Srf in controlling the organization of actin cytoskeleton and actin-based protrusions for myoblast fusion in mammals and its requirement to achieve efficient hypertrophic myofiber growth.
Collapse
Affiliation(s)
- Voahangy Randrianarison-Huetz
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Aikaterini Papaefthymiou
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Gaëlle Herledan
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Chiara Noviello
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Ulduz Faradova
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | | | - Alessandra Pincini
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Emilie Schol
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Jean François Decaux
- Université Pierre et Marie Curie Paris 6, Centre National de la Recherche Scientifique UMR8256, Institut National de la Santé et de la Recherche Médicale U1164, Institute of Biology Paris-Seine, Paris, France
| | - Pascal Maire
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France.,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| | - Stéphane Vassilopoulos
- Institut National de la Santé et de la Recherche Médicale/University Pierre and Marie Curie UMR-S974, Institut de Myologie, Paris, France
| | - Athanassia Sotiropoulos
- Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France .,Centre National de la Recherche Scientifique UMR8104, Paris, France.,Université Paris Descartes, Paris, France
| |
Collapse
|
194
|
de Perini A, Dimauro I, Duranti G, Fantini C, Mercatelli N, Ceci R, Di Luigi L, Sabatini S, Caporossi D. The p75 NTR-mediated effect of nerve growth factor in L6C5 myogenic cells. BMC Res Notes 2017; 10:686. [PMID: 29202822 PMCID: PMC5716223 DOI: 10.1186/s13104-017-2994-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/25/2017] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE During muscle development or regeneration, myocytes produce nerve growth factor (NGF) as well as its tyrosine-kinase and p75-neurotrophin (p75NTR) receptors. It has been published that the p75NTR receptor could represent a key regulator of NGF-mediated myoprotective effect on satellite cells, but the precise function of NGF/p75 signaling pathway on myogenic cell proliferation, survival and differentiation remains fragmented and controversial. Here, we verified the role of NGF in the growth, survival and differentiation of p75NTR-expressing L6C5 myogenic cells, specifically inquiring for the putative involvement of the nuclear factor κB (NFκB) and the small heat shock proteins (sHSPs) αB-crystallin and Hsp27 in these processes. RESULTS Although NGF was not effective in modulating myogenic cell growth or survival in both standard or stress conditions, we demonstrated for the first time that, under serum deprivation, NGF sustained the activity of some key enzymes involved in energy metabolism. Moreover, we confirmed that NGF promotes myogenic fusion and expression of the structural protein myosin heavy chain while modulating NFκB activation and the content of sHSPs correlated with the differentiation process. We conclude that p75NTR is sufficient to mediate the modulation of L6C5 myogenic differentiation by NGF in term of structural, metabolic and functional changes.
Collapse
Affiliation(s)
- Alessandra de Perini
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| | - Ivan Dimauro
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| | - Guglielmo Duranti
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| | - Cristina Fantini
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| | - Neri Mercatelli
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
- Laboratory of Cellular and Molecular Neurobiology, CERC, Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Roberta Ceci
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| | - Luigi Di Luigi
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| | - Stefania Sabatini
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| | - Daniela Caporossi
- Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis, 15, 00135 Rome, Italy
| |
Collapse
|
195
|
Insights into the Pathogenic Secondary Symptoms Caused by the Primary Loss of Dystrophin. J Funct Morphol Kinesiol 2017. [DOI: 10.3390/jfmk2040044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
196
|
The histone code reader Spin1 controls skeletal muscle development. Cell Death Dis 2017; 8:e3173. [PMID: 29168801 PMCID: PMC5775400 DOI: 10.1038/cddis.2017.468] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/12/2017] [Accepted: 07/28/2017] [Indexed: 01/06/2023]
Abstract
While several studies correlated increased expression of the histone code reader Spin1 with tumor formation or growth, little is known about physiological functions of the protein. We generated Spin1M5 mice with ablation of Spin1 in myoblast precursors using the Myf5-Cre deleter strain. Most Spin1M5 mice die shortly after birth displaying severe sarcomere disorganization and necrosis. Surviving Spin1M5 mice are growth-retarded and exhibit the most prominent defects in soleus, tibialis anterior, and diaphragm muscle. Transcriptome analyses of limb muscle at embryonic day (E) 15.5, E16.5, and at three weeks of age provided evidence for aberrant fetal myogenesis and identified deregulated skeletal muscle (SkM) functional networks. Determination of genome-wide chromatin occupancy in primary myoblast revealed direct Spin1 target genes and suggested that deregulated basic helix-loop-helix transcription factor networks account for developmental defects in Spin1M5 fetuses. Furthermore, correlating histological and transcriptome analyses, we show that aberrant expression of titin-associated proteins, abnormal glycogen metabolism, and neuromuscular junction defects contribute to SkM pathology in Spin1M5 mice. Together, we describe the first example of a histone code reader controlling SkM development in mice, which hints at Spin1 as a potential player in human SkM disease.
Collapse
|
197
|
Lack of Heme Oxygenase-1 Induces Inflammatory Reaction and Proliferation of Muscle Satellite Cells after Cardiotoxin-Induced Skeletal Muscle Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:491-506. [PMID: 29169990 DOI: 10.1016/j.ajpath.2017.10.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/12/2017] [Accepted: 10/23/2017] [Indexed: 12/29/2022]
Abstract
Heme oxygenase-1 (HO-1, Hmox1) regulates viability, proliferation, and differentiation of many cell types; hence, it may affect regeneration of injured skeletal muscle. Here, we injected cardiotoxin into gastrocnemius muscle of Hmox1+/+ and Hmox1-/- animals and analyzed cellular response after muscle injury, focusing on muscle satellite cells (SCs), inflammatory reaction, fibrosis, and formation of new blood vessels. HO-1 is strongly induced after muscle injury, being expressed mostly in the infiltrating leukocytes (CD45+ cells), including macrophages (F4/80+ cells). Lack of HO-1 augments skeletal muscle injury, evidenced by increased creatinine kinase and lactate dehydrogenase, as well as expression of monocyte chemoattractant protein-1, IL-6, IL-1β, and insulin-like growth factor-1. This, together with disturbed proportion of M1/M2 macrophages, accompanied by enhanced formation of arterioles, may be responsible for shift of Hmox1-/- myofiber size distribution toward larger one. Importantly, HO-1-deficient SCs are prone to activation and have higher proliferation on injury. This effect can be partially mimicked by stimulation of Hmox1+/+ SCs with monocyte chemoattractant protein-1, IL-6, IL-1β, and is associated with increased MyoD expression, suggesting that Hmox1-/- SCs are shifted toward more differentiated myogenic population. However, multiple rounds of degeneration/regeneration in conditions of HO-1 deficiency may lead to exhaustion of SC pool, and the number of SCs is decreased in old Hmox1-/- mice. In summary, HO-1 modulates muscle repair mechanisms preventing its uncontrolled acceleration.
Collapse
|
198
|
Yue F, Bi P, Wang C, Li J, Liu X, Kuang S. Conditional Loss of Pten in Myogenic Progenitors Leads to Postnatal Skeletal Muscle Hypertrophy but Age-Dependent Exhaustion of Satellite Cells. Cell Rep 2017; 17:2340-2353. [PMID: 27880908 DOI: 10.1016/j.celrep.2016.11.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/19/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle stem cells (satellite cells [SCs]) are normally maintained in a quiescent (G0) state. Muscle injury not only activates SCs locally, but also alerts SCs in distant uninjured muscles via circulating factors. The resulting GAlert SCs are adapted to regenerative cues and regenerate injured muscles more efficiently, but whether they provide any long-term benefits to SCs is unknown. Here, we report that embryonic myogenic progenitors lacking the phosphatase and tensin homolog (Pten) exhibit enhanced proliferation and differentiation, resulting in muscle hypertrophy but fewer SCs in adult muscles. Interestingly, Pten null SCs are predominantly in the GAlert state, even in the absence of an injury. The GAlert SCs are deficient in self-renewal and subjected to accelerated depletion during regeneration and aging and fail to repair muscle injury in old mice. Our findings demonstrate a key requirement of Pten in G0 entry of SCs and provide functional evidence that prolonged GAlert leads to stem cell depletion and regenerative failure.
Collapse
Affiliation(s)
- Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Pengpeng Bi
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Chao Wang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jie Li
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Xiaoqi Liu
- Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
199
|
Tatapudy S, Aloisio F, Barber D, Nystul T. Cell fate decisions: emerging roles for metabolic signals and cell morphology. EMBO Rep 2017; 18:2105-2118. [PMID: 29158350 DOI: 10.15252/embr.201744816] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/14/2017] [Accepted: 10/24/2017] [Indexed: 12/25/2022] Open
Abstract
Understanding how cell fate decisions are regulated is a fundamental goal of developmental and stem cell biology. Most studies on the control of cell fate decisions address the contributions of changes in transcriptional programming, epigenetic modifications, and biochemical differentiation cues. However, recent studies have found that other aspects of cell biology also make important contributions to regulating cell fate decisions. These cues can have a permissive or instructive role and are integrated into the larger network of signaling, functioning both upstream and downstream of developmental signaling pathways. Here, we summarize recent insights into how cell fate decisions are influenced by four aspects of cell biology: metabolism, reactive oxygen species (ROS), intracellular pH (pHi), and cell morphology. For each topic, we discuss how these cell biological cues interact with each other and with protein-based mechanisms for changing gene transcription. In addition, we highlight several questions that remain unanswered in these exciting and relatively new areas of the field.
Collapse
Affiliation(s)
- Sumitra Tatapudy
- Departments of Anatomy and OB-GYN/RS, University of California, San Francisco, San Francisco, CA, USA
| | - Francesca Aloisio
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Diane Barber
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA, USA
| | - Todd Nystul
- Departments of Anatomy and OB-GYN/RS, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
200
|
Pease LI, Clegg PD, Proctor CJ, Shanley DJ, Cockell SJ, Peffers MJ. Cross platform analysis of transcriptomic data identifies ageing has distinct and opposite effects on tendon in males and females. Sci Rep 2017; 7:14443. [PMID: 29089527 PMCID: PMC5663855 DOI: 10.1038/s41598-017-14650-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/13/2017] [Indexed: 01/21/2023] Open
Abstract
The development of tendinopathy is influenced by a variety of factors including age, gender, sex hormones and diabetes status. Cross platform comparative analysis of transcriptomic data elucidated the connections between these entities in the context of ageing. Tissue-engineered tendons differentiated from bone marrow derived mesenchymal stem cells from young (20-24 years) and old (54-70 years) donors were assayed using ribonucleic acid sequencing (RNA-seq). Extension of the experiment to microarray and RNA-seq data from tendon identified gender specific gene expression changes highlighting disparity with existing literature and published pathways. Separation of RNA-seq data by sex revealed underlying negative binomial distributions which increased statistical power. Sex specific de novo transcriptome assemblies generated fewer larger transcripts that contained miRNAs, lincRNAs and snoRNAs. The results identify that in old males decreased expression of CRABP2 leads to cell proliferation, whereas in old females it leads to cellular senescence. In conjunction with existing literature the results explain gender disparity in the development and types of degenerative diseases as well as highlighting a wide range of considerations for the analysis of transcriptomic data. Wider implications are that degenerative diseases may need to be treated differently in males and females because alternative mechanisms may be involved.
Collapse
Affiliation(s)
- Louise I Pease
- MRC - Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool, UK
| | - Peter D Clegg
- MRC - Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool, UK
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, The University of Liverpool, Leahurst Campus, Neston, CH64 7TE, UK
| | - Carole J Proctor
- MRC - Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool, UK
- Institute of Cellular Medicine, Newcastle University, Newcastle, NE2 4HH, UK
| | - Daryl J Shanley
- MRC - Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool, UK
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle, NE1 7RU, UK
| | - Simon J Cockell
- Faculty of Medical Sciences, Bioinformatics Support Unit, Framlington Place, Newcastle University, Newcastle, NE2 4HH, UK
| | - Mandy J Peffers
- MRC - Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing (CIMA), Liverpool, UK.
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, The University of Liverpool, Leahurst Campus, Neston, CH64 7TE, UK.
| |
Collapse
|