51
|
Olson LC, Redden JT, Schwartz Z, Cohen DJ, McClure MJ. Advanced Glycation End-Products in Skeletal Muscle Aging. Bioengineering (Basel) 2021; 8:bioengineering8110168. [PMID: 34821734 PMCID: PMC8614898 DOI: 10.3390/bioengineering8110168] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/17/2022] Open
Abstract
Advanced age causes skeletal muscle to undergo deleterious changes including muscle atrophy, fast-to-slow muscle fiber transition, and an increase in collagenous material that culminates in the age-dependent muscle wasting disease known as sarcopenia. Advanced glycation end-products (AGEs) non-enzymatically accumulate on the muscular collagens in old age via the Maillard reaction, potentiating the accumulation of intramuscular collagen and stiffening the microenvironment through collagen cross-linking. This review contextualizes known aspects of skeletal muscle extracellular matrix (ECM) aging, especially the role of collagens and AGE cross-linking, and underpins the motor nerve’s role in this aging process. Specific directions for future research are also discussed, with the understudied role of AGEs in skeletal muscle aging highlighted. Despite more than a half century of research, the role that intramuscular collagen aggregation and cross-linking plays in sarcopenia is well accepted yet not well integrated with current knowledge of AGE’s effects on muscle physiology. Furthermore, the possible impact that motor nerve aging has on intramuscular cross-linking and muscular AGE levels is posited.
Collapse
Affiliation(s)
- Lucas C. Olson
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
- Department of Gerontology, College of Health Professions, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - James T. Redden
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - David J. Cohen
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
| | - Michael J. McClure
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; (L.C.O.); (J.T.R.); (Z.S.); (D.J.C.)
- Correspondence:
| |
Collapse
|
52
|
Buonaiuto G, Desideri F, Taliani V, Ballarino M. Muscle Regeneration and RNA: New Perspectives for Ancient Molecules. Cells 2021; 10:cells10102512. [PMID: 34685492 PMCID: PMC8533951 DOI: 10.3390/cells10102512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022] Open
Abstract
The ability of the ribonucleic acid (RNA) to self-replicate, combined with a unique cocktail of chemical properties, suggested the existence of an RNA world at the origin of life. Nowadays, this hypothesis is supported by innovative high-throughput and biochemical approaches, which definitively revealed the essential contribution of RNA-mediated mechanisms to the regulation of fundamental processes of life. With the recent development of SARS-CoV-2 mRNA-based vaccines, the potential of RNA as a therapeutic tool has received public attention. Due to its intrinsic single-stranded nature and the ease with which it is synthesized in vitro, RNA indeed represents the most suitable tool for the development of drugs encompassing every type of human pathology. The maximum effectiveness and biochemical versatility is achieved in the guise of non-coding RNAs (ncRNAs), which are emerging as multifaceted regulators of tissue specification and homeostasis. Here, we report examples of coding and ncRNAs involved in muscle regeneration and discuss their potential as therapeutic tools. Small ncRNAs, such as miRNA and siRNA, have been successfully applied in the treatment of several diseases. The use of longer molecules, such as lncRNA and circRNA, is less advanced. However, based on the peculiar properties discussed below, they represent an innovative pool of RNA biomarkers and possible targets of clinical value.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- COVID-19
- Homeostasis
- Humans
- Mice
- MicroRNAs/metabolism
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/virology
- Myocardium/metabolism
- Origin of Life
- RNA, Circular
- RNA, Long Noncoding/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- RNA, Small Untranslated/genetics
- RNA, Untranslated/genetics
- RNA, Viral/metabolism
- Regeneration
- SARS-CoV-2/genetics
Collapse
Affiliation(s)
- Giulia Buonaiuto
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
| | - Fabio Desideri
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
- Center for Life Nano & Neuro-Science of Instituto Italiano di Tecnologia (IIT), 00161 Rome, Italy
| | - Valeria Taliani
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
| | - Monica Ballarino
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (G.B.); (F.D.); (V.T.)
- Correspondence:
| |
Collapse
|
53
|
de Souza C, Eyng C, Viott A, de Avila A, Pacheco W, Junior N, Kohler T, Tenorio K, Cirilo E, Nunes R. Effect of dietary guanidinoacetic acid or nucleotides supplementation on growth performances, carcass traits, meat quality and occurrence of myopathies in broilers. Livest Sci 2021. [DOI: 10.1016/j.livsci.2021.104659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
54
|
Ramani S, Ko D, Kim B, Cho C, Kim W, Jo C, Lee CK, Kang J, Hur S, Park S. Technical requirements for cultured meat production: a review. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2021; 63:681-692. [PMID: 34447948 PMCID: PMC8367405 DOI: 10.5187/jast.2021.e45] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/28/2022]
Abstract
Environment, food, and disease have a selective force on the present and future
as well as our genome. Adaptation of livestock and the environmental nexus,
including forest encroachment for anthropological needs, has been proven to
cause emerging infectious diseases. Further, these demand changes in meat
production and market systems. Meat is a reliable source of protein, with a
majority of the world population consumes meat. To meet the increasing demands
of meat production as well as address issues, such as current environmental
pollution, animal welfare, and outbreaks, cellular agriculture has emerged as
one of the next industrial revolutions. Lab grown meat or cell cultured meat is
a promising way to pursue this; however, it still needs to resemble traditional
meat and be assured safety for human consumption. Further, to mimic the
palatability of traditional meat, the process of cultured meat production starts
from skeletal muscle progenitor cells isolated from animals that proliferate and
differentiate into skeletal muscle using cell culture techniques. Due to several
lacunae in the current approaches, production of muscle replicas is not possible
yet. Our review shows that constant research in this field will resolve the
existing constraints and enable successful cultured meat production in the near
future. Therefore, production of cultured meat is a better solution that looks
after environmental issues, spread of outbreaks, antibiotic resistance through
the zoonotic spread, food and economic crises.
Collapse
Affiliation(s)
| | - Deunsol Ko
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Bosung Kim
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Changjun Cho
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Woosang Kim
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | | | - Sunjin Hur
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Sungkwon Park
- Department of Food Science and Biotechnology, Sejong University, Seoul 05006, Korea
| |
Collapse
|
55
|
Khuu S, Fernandez JW, Handsfield GG. A Coupled Mechanobiological Model of Muscle Regeneration In Cerebral Palsy. Front Bioeng Biotechnol 2021; 9:689714. [PMID: 34513808 PMCID: PMC8429491 DOI: 10.3389/fbioe.2021.689714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/06/2021] [Indexed: 01/05/2023] Open
Abstract
Cerebral palsy is a neuromusculoskeletal disorder associated with muscle weakness, altered muscle architecture, and progressive musculoskeletal symptoms that worsen with age. Pathological changes at the level of the whole muscle have been shown; however, it is unclear why this progression of muscle impairment occurs at the cellular level. The process of muscle regeneration is complex, and the interactions between cells in the muscle milieu should be considered in the context of cerebral palsy. In this work, we built a coupled mechanobiological model of muscle damage and regeneration to explore the process of muscle regeneration in typical and cerebral palsy conditions, and whether a reduced number of satellite cells in the cerebral palsy muscle environment could cause the muscle regeneration cycle to lead to progressive degeneration of muscle. The coupled model consisted of a finite element model of a muscle fiber bundle undergoing eccentric contraction, and an agent-based model of muscle regeneration incorporating satellite cells, inflammatory cells, muscle fibers, extracellular matrix, fibroblasts, and secreted cytokines. Our coupled model simulated damage from eccentric contraction followed by 28 days of regeneration within the muscle. We simulated cyclic damage and regeneration for both cerebral palsy and typically developing muscle milieus. Here we show the nonlinear effects of altered satellite cell numbers on muscle regeneration, where muscle repair is relatively insensitive to satellite cell concentration above a threshold, but relatively sensitive below that threshold. With the coupled model, we show that the fiber bundle geometry undergoes atrophy and fibrosis with too few satellite cells and excess extracellular matrix, representative of the progression of cerebral palsy in muscle. This work uses in silico modeling to demonstrate how muscle degeneration in cerebral palsy may arise from the process of cellular regeneration and a reduced number of satellite cells.
Collapse
Affiliation(s)
- Stephanie Khuu
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Justin W. Fernandez
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | | |
Collapse
|
56
|
Fujikura Y, Sugihara H, Hatakeyama M, Oishi K, Yamanouchi K. Ketogenic diet with medium-chain triglycerides restores skeletal muscle function and pathology in a rat model of Duchenne muscular dystrophy. FASEB J 2021; 35:e21861. [PMID: 34416029 DOI: 10.1096/fj.202100629r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/16/2021] [Accepted: 08/04/2021] [Indexed: 12/28/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an intractable genetic disease associated with progressive skeletal muscle weakness and degeneration. Recently, it was reported that intraperitoneal injections of ketone bodies partially ameliorated muscular dystrophy by increasing satellite cell (SC) proliferation. Here, we evaluated whether a ketogenic diet (KD) with medium-chain triglycerides (MCT-KD) could alter genetically mutated DMD in model rats. We found that the MCT-KD significantly increased muscle strength and fiber diameter in these rats. The MCT-KD significantly suppressed the key features of DMD, namely, muscle necrosis, inflammation, and subsequent fibrosis. Immunocytochemical analysis revealed that the MCT-KD promoted the proliferation of muscle SCs, suggesting enhanced muscle regeneration. The muscle strength of DMD model rats fed with MCT-KD was significantly improved even at the age of 9 months. Our findings suggested that the MCT-KD ameliorates muscular dystrophy by inhibiting myonecrosis and promoting the proliferation of muscle SCs. As far as we can ascertain, this is the first study to apply a functional diet as therapy for DMD in experimental animals. Further studies are needed to elucidate the underlying mechanisms of the MCT-KD-induced improvement of DMD.
Collapse
Affiliation(s)
- Yuri Fujikura
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Hidetoshi Sugihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | | | - Katsutaka Oishi
- Healthy Food Science Research Group, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan.,Department of Applied Biological Science, Graduate School of Science and Technology, Tokyo University of Science, Noda, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan.,School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| |
Collapse
|
57
|
Song R, Zhao S, Xu Y, Hu J, Ke S, Li F, Tian G, Zheng X, Li J, Gu L, Xu Y. MRTF-A regulates myoblast commitment to differentiation by targeting PAX7 during muscle regeneration. J Cell Mol Med 2021; 25:8645-8661. [PMID: 34347392 PMCID: PMC8435411 DOI: 10.1111/jcmm.16820] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Myocardin-related transcription factor-A/serum response factor (MRTF-A/SRF), a well-known transcriptional programme, has been proposed to play crucial roles in skeletal muscle development and function. However, whether MRTF-A participates in muscle regeneration and the molecular mechanisms are not completely understood. Here, we show that MRTF-A levels are highly correlated with myogenic genes using a RNA-seq assay, which reveal that MRTF-A knockdown in C2C12 cells significantly reduces PAX7 expression. Subsequent in vitro and in vivo data show that MRTF-A and PAX7 present identical expression patterns during myoblast differentiation and CTX-induced muscle injury and repair. Remarkably, MRTF-A overexpression promotes myoblast proliferation, while inhibiting cell differentiation and the expression of MyoD and MyoG. MRTF-A loss of function produces the opposite effect. Moreover, mice with lentivirus (MRTF-A) injection possesses more PAX7+ satellite cells, but less differentiating MyoD+ and MyoG+ cells, leading subsequently to diminished muscle regeneration. Our mechanistic results reveal that MRTF-A contributes to PAX7-mediated myoblast self-renewal, proliferation, and differentiation by binding to its distal CArG box. Overall, we propose that MRTF-A functions as a novel PAX7 regulator upon myoblast commitment to differentiation, which could provide pathways for dictating muscle stem cell fate and open new avenues to explore stem cell-based therapy for muscle degenerative diseases.
Collapse
Affiliation(s)
- Ruhui Song
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Shengnan Zhao
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Yue Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Jian Hu
- Animal Disease Control and Prevention Centre of Chongqing City, Chongqing, 401120, China
| | - Shuangao Ke
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Fan Li
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Gaohui Tian
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Xiao Zheng
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Jiajun Li
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Lixing Gu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| | - Yao Xu
- Institute of Biology and Medicine, College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, 430081, China
| |
Collapse
|
58
|
Rosa I, Marini M, Manetti M. Telocytes: An Emerging Component of Stem Cell Niche Microenvironment. J Histochem Cytochem 2021; 69:795-818. [PMID: 34165348 DOI: 10.1369/00221554211025489] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Telocytes (TCs) are newly identified interstitial cells characterized by thin and long cytoplasmic processes, called telopodes, which exhibit a distinctive moniliform shape and, often, a sinuous trajectory. Telopodes typically organize in intricate networks within the stromal space of most organs, where they communicate with neighboring cells by means of specialized cell-to-cell junctions or shedding extracellular vesicles. Hence, TCs are generally regarded as supporting cells that help in the maintenance of local tissue homeostasis, with an ever-increasing number of studies trying to explore their functions both in physiological and pathological conditions. Notably, TCs appear to be part of stem cell (SC) niches in different organs, including the intestine, skeletal muscle, heart, lung, and skin. Indeed, growing evidence points toward a possible implication of TCs in the regulation of the activity of tissue-resident SCs and in shaping the SC niche microenvironment, thus contributing to tissue renewal and repair. Here, we review how the introduction of TCs into the scientific literature has deepened our knowledge of the stromal architecture focusing on the intestine and skeletal muscle, two organs in which the recently unveiled unique relationship between TCs and SCs is currently in the spotlight as potential target for tissue regenerative purposes.
Collapse
Affiliation(s)
- Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mirca Marini
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
59
|
In vivo partial reprogramming of myofibers promotes muscle regeneration by remodeling the stem cell niche. Nat Commun 2021; 12:3094. [PMID: 34035273 PMCID: PMC8149870 DOI: 10.1038/s41467-021-23353-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 04/22/2021] [Indexed: 01/09/2023] Open
Abstract
Short-term, systemic expression of the Yamanaka reprogramming factors (Oct-3/4, Sox2, Klf4 and c-Myc [OSKM]) has been shown to rejuvenate aging cells and promote tissue regeneration in vivo. However, the mechanisms by which OSKM promotes tissue regeneration are unknown. In this work, we focus on a specific tissue and demonstrate that local expression of OSKM, specifically in myofibers, induces the activation of muscle stem cells or satellite cells (SCs), which accelerates muscle regeneration in young mice. In contrast, expressing OSKM directly in SCs does not improve muscle regeneration. Mechanistically, expressing OSKM in myofibers regulates the expression of genes important for the SC microenvironment, including upregulation of p21, which in turn downregulates Wnt4. This is critical because Wnt4 is secreted by myofibers to maintain SC quiescence. Thus, short-term induction of the Yamanaka factors in myofibers may promote tissue regeneration by modifying the stem cell niche. Short term systemic expression of the reprogramming factors Oct-3/4, Sox2, Klf4, c-Myc (OSKM) rejuvenates aging cells and promotes tissue regeneration. Here the authors show that myofiber-specific expression of OSKM accelerates muscle regeneration by reducing secretion of muscle stem cell quiescence promoting Wnt4.
Collapse
|
60
|
Ludikhuize MC, Rodríguez Colman MJ. Metabolic Regulation of Stem Cells and Differentiation: A Forkhead Box O Transcription Factor Perspective. Antioxid Redox Signal 2021; 34:1004-1024. [PMID: 32847377 DOI: 10.1089/ars.2020.8126] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Stem cell activation and differentiation occur along changes in cellular metabolism. Metabolic transitions translate into changes in redox balance, cell signaling, and epigenetics, thereby regulating these processes. Metabolic transitions are key regulators of cell fate and exemplify the moonlighting nature of many metabolic enzymes and their associated metabolites. Recent Advances: Forkhead box O transcription factors (FOXOs) are bona fide regulators of cellular homeostasis. FOXOs are multitasking proteins able to regulate cell cycle, cellular metabolism, and redox state. Recent and ongoing research poses FOXOs as key factors in stem cell maintenance and differentiation in several tissues. Critical Issues: The multitasking nature of FOXOs and their tissue-specific expression patterns hinders to disclose a possible conserved mechanism of regulation of stem cell maintenance and differentiation. Moreover, cellular metabolism, cell signaling, and epigenetics establish complex regulatory interactions, which challenge the establishment of the causal/temporal nature of metabolic changes and stem cell activation and differentiation. Future Directions: The development of single-cell technologies and in vitro models able to reproduce the dynamics of stem cell differentiation are actively contributing to define the role of metabolism in this process. This knowledge is key to understanding and designing therapies for those pathologies where the balance between proliferation and differentiation is lost. Importantly, metabolic interventions could be applied to optimize stem cell cultures meant for therapeutical applications, such as transplantations, to treat autoimmune and degenerative disorders. Antioxid. Redox Signal. 34, 1004-1024.
Collapse
Affiliation(s)
- Marlies Corine Ludikhuize
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - María José Rodríguez Colman
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
61
|
Kim H, Jeong JH, Fendereski M, Lee HS, Kang DY, Hur SS, Amirian J, Kim Y, Pham NT, Suh N, Hwang NSY, Ryu S, Yoon JK, Hwang Y. Heparin-Mimicking Polymer-Based In Vitro Platform Recapitulates In Vivo Muscle Atrophy Phenotypes. Int J Mol Sci 2021; 22:ijms22052488. [PMID: 33801235 PMCID: PMC7957884 DOI: 10.3390/ijms22052488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
The cell–cell/cell–matrix interactions between myoblasts and their extracellular microenvironment have been shown to play a crucial role in the regulation of in vitro myogenic differentiation and in vivo skeletal muscle regeneration. In this study, by harnessing the heparin-mimicking polymer, poly(sodium-4-styrenesulfonate) (PSS), which has a negatively charged surface, we engineered an in vitro cell culture platform for the purpose of recapitulating in vivo muscle atrophy-like phenotypes. Our initial findings showed that heparin-mimicking moieties inhibited the fusion of mononucleated myoblasts into multinucleated myotubes, as indicated by the decreased gene and protein expression levels of myogenic factors, myotube fusion-related markers, and focal adhesion kinase (FAK). We further elucidated the underlying molecular mechanism via transcriptome analyses, observing that the insulin/PI3K/mTOR and Wnt signaling pathways were significantly downregulated by heparin-mimicking moieties through the inhibition of FAK/Cav3. Taken together, the easy-to-adapt heparin-mimicking polymer-based in vitro cell culture platform could be an attractive platform for potential applications in drug screening, providing clear readouts of changes in insulin/PI3K/mTOR and Wnt signaling pathways.
Collapse
Affiliation(s)
- Hyunbum Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Mona Fendereski
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Hyo-Shin Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Da Yeon Kang
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Sung Sik Hur
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
| | - Jhaleh Amirian
- Institute of Tissue Regeneration, Soonchunhyang University, Asan-si 31538, Korea;
| | - Yunhye Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nghia Thi Pham
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nayoung Suh
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Nathaniel Suk-Yeon Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| |
Collapse
|
62
|
Sarkar A, Saha S, Paul A, Maji A, Roy P, Maity TK. Understanding stem cells and its pivotal role in regenerative medicine. Life Sci 2021; 273:119270. [PMID: 33640402 DOI: 10.1016/j.lfs.2021.119270] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/06/2021] [Accepted: 02/14/2021] [Indexed: 02/07/2023]
Abstract
Stem cells (SCs) are clonogenic cells that develop into the specialized cells which later responsible for making up various types of tissue in the human body. SCs are not only the appropriate source of information for cell division, molecular and cellular processes, and tissue homeostasis but also one of the major putative biological aids to diagnose and cure various degenerative diseases. This study emphasises on various research outputs that occurred in the past two decades. This will give brief information on classification, differentiation, detection, and various isolation techniques of SCs. Here, the various signalling pathways which includes WNT, Sonic hedgehog, Notch, BMI1 and C-met pathways and how does it effect on the regeneration of various classes of SCs and factors that regulates the potency of the SCs are also been discussed. We also focused on the application of SCs in the area of regenerative medicine along with the cellular markers that are useful as salient diagnostic or curative tools or in both, by the process of reprogramming, which includes diabetes, cancer, cardiovascular disorders and neurological disorders. The biomarkers that are mentioned in various literatures and experiments include PDX1, FOXA2, HNF6, and NKX6-1 (for diabetes); CD33, CD24, CD133 (for cancer); c-Kit, SCA-1, Wilm's tumor 1 (for cardiovascular disorders); and OCT4, SOX2, c-MYC, EN1, DAT and VMAT2 (for neurological disorders). In this review, we come to know the advancements and scopes of potential SC-based therapies, its diverse applications in clinical fields that can be helpful in the near future.
Collapse
Affiliation(s)
- Arnab Sarkar
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Sanjukta Saha
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Abhik Paul
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Avik Maji
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Puspita Roy
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India
| | - Tapan Kumar Maity
- Department of Pharmaceutical Technology, Jadavpur University, West Bengal, Kolkata 700032, India.
| |
Collapse
|
63
|
Gudagudi KB, Myburgh KH. Methods to Mimic In Vivo Muscle Cell Biology in Primary Human Myoblasts Using Quiescence as a Guidepost in Regenerative Medicine Research. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:176-189. [PMID: 33635139 DOI: 10.1089/omi.2020.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Regenerative medicine research and testing of new therapeutics for muscle-related human diseases call for a deeper understanding of how human myoblasts gain and maintain quiescence in vitro versus in vivo. The more closely we can experimentally simulate the in vivo environment, the more relevance in vitro research on myoblasts will have. In this context, isolation of satellite cells from muscle tissue causes activation while myoblasts remain activated in culture, thus not simulating quiescence as in their in vivo niche. Cells synchronized for cell cycle present a good starting point for experimental intervention. In the past, myoblast quiescence has been induced using suspension culture (SuCu) and, recently, by knockout serum replacement (KOSR)-supplemented culture media. We assessed the proportion of cells in G0 and molecular regulators after combining the two quiescence-inducing approaches. Quiescence was induced in primary human myoblasts (PHMs) in vitro using KOSR-treatment for 10 days or suspension in viscous media for 2 days (SuCu), or suspension combined with KOSR-treatment for 2 days (blended method, SuCu-KOSR). Quiescence and synchronization were achieved with all three protocols (G0/G1 cell cycle arrest >90% cells). Fold-change of cell cycle controller p21 mRNA for KOSR and SuCu was 3.23 ± 0.30 and 2.86 ± 0.15, respectively. Since this was already a significant change (p < 0.05), no further change was gained with the blended method. But SuCu-KOSR significantly decreased Ki67 (p = 0.0019). Myogenic regulatory factors, Myf5 and MyoD gene expression in PHMs were much more suppressed (p = 0.0004 and p = 0.0034, respectively) in SuCu-KOSR, compared to SuCu alone. In conclusion, a homogenous pool of quiescent primary myoblasts synchronized in the G0 cell cycle phase was achieved with cells from three different donors regardless of the experimental protocol. Myogenic dedifferentiation at the level of Myogenic Regulatory Factors was greater when exposed to the blend of suspension and serum-free culture. We suggest that this blended new protocol can be considered in future biomedical research if differentiation is detected too early during myoblast expansion. This shall also inform new ways to bridge the in vitro and in vivo divides in regenerative medicine research.
Collapse
Affiliation(s)
- Kirankumar B Gudagudi
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Kathryn H Myburgh
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
64
|
Ramalho L, Nedjari S, Guarino R, Awaja F, Gugutkov D, Altankov G. Fibronectin/thermo-responsive polymer scaffold as a dynamic ex vivo niche for mesenchymal stem cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:129. [PMID: 33252710 DOI: 10.1007/s10856-020-06461-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 10/31/2020] [Indexed: 06/12/2023]
Abstract
In this paper, we created a dynamic adhesive environment (DAE) for adipose tissue-derived mesenchymal stem cells (ADMSCs) cultured on smart thermo-responsive substrates, i.e., poly (N-isopropyl acrylamide) (PNIPAM), via introducing periodic changes in the culture temperature. We further explored the particular role of adsorbed fibronectin (FN), an important cell adhesive protein that was recently attributed to the recruitment of stem cells in the niche. The engineered FN/PNIPAM DAE system significantly increased the symmetric renewal of ADMSCs, particularly between passages 7 and 9 (p7-p9), before it dropped down to the level of the control (FN-coated TC polystyrene). This decline in the growth curve was consistent with the increased number of senescent cells, the augmented average cell size and the suppressed FN matrix secretion at late passages (p10-p12), all of them characteristic for stem cells ageing, which equivocally tended to slow down at our DAE system. FN supported also the osteogenic response of ADMSCs (apart from the previous observations with plain PNIPAM substrata) indicated by the significant increase of alkaline phosphatase (ALP) activity at days 7 and 14. The minimal changes in the Ca deposition, however, suggest a restricted effect of DAE on the early osteogenic response of ADMSCs only. Thus, the engineering of niche-like DAE involving FN uncovers a new tissue engineering strategy for gaining larger amounts of functionally active stem cells for clinical application.
Collapse
Affiliation(s)
- Laura Ramalho
- ICREA, Barcelona, Spain
- Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | | | - Roberto Guarino
- École Polytechnique Fédérale de Lausanne (EPFL), Swiss Plasma Center (SPC), CH-5232, Villigen PSI, Switzerland
| | - Firas Awaja
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
- Engmat Ltd., Clybaun Road, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) and Centre for Research in Medical Devices (CÚRAM) at National University of Ireland, Galway, Ireland
| | | | - George Altankov
- ICREA, Barcelona, Spain.
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.
- Associate Member Institute for Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| |
Collapse
|
65
|
Zembron-Lacny A, Morawin B, Wawrzyniak-Gramacka E, Gramacki J, Jarmuzek P, Kotlega D, Ziemann E. Multiple Cryotherapy Attenuates Oxi-Inflammatory Response Following Skeletal Muscle Injury. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17217855. [PMID: 33120891 PMCID: PMC7663269 DOI: 10.3390/ijerph17217855] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023]
Abstract
The oxi-inflammatory response is part of the natural process mobilizing leukocytes and satellite cells that contribute to clearance and regeneration of damaged muscle tissue. In sports medicine, a number of post-injury recovery strategies, such as whole-body cryotherapy (WBC), are used to improve skeletal muscle regeneration often without scientific evidence of their benefits. The study was designed to assess the impact of WBC on circulating mediators of skeletal muscle regeneration. Twenty elite athletes were randomized to WBC group (3-min exposure to −120 °C, twice a day for 7 days) and control group. Blood samples were collected before the first WBC session and 1 day after the last cryotherapy exposure. WBC did not affect the indirect markers of muscle damage but significantly reduced the generation of reactive oxygen and nitrogen species (H2O2 and NO) as well as the concentrations of serum interleukin 1β (IL-1β) and C-reactive protein (CRP). The changes in circulating growth factors, hepatocyte growth factor (HGF), insulin-like growth factor (IGF-1), platelet-derived growth factor (PDGFBB), vascular endothelial growth factor (VEGF), and brain-derived neurotrophic factor (BDNF), were also reduced by WBC exposure. The study demonstrated that WBC attenuates the cascade of injury–repair–regeneration of skeletal muscles whereby it may delay skeletal muscle regeneration.
Collapse
Affiliation(s)
- Agnieszka Zembron-Lacny
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland; (B.M.); (E.W.-G.)
- Correspondence: ; Tel.: +48-50267-4130
| | - Barbara Morawin
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland; (B.M.); (E.W.-G.)
| | - Edyta Wawrzyniak-Gramacka
- Department of Applied and Clinical Physiology, Collegium Medicum University of Zielona Gora, 65-417 Zielona Gora, Poland; (B.M.); (E.W.-G.)
| | - Jaroslaw Gramacki
- Centre of Information Technologies, University of Zielona Gora, 65-417 Zielona Gora, Poland;
| | - Pawel Jarmuzek
- Department of Nervous System Diseases, Collegium Medium University of Zielona Gora, Neurosurgery Center University Hospital in Zielona Gora, 65-417 Zielona Gora, Poland;
| | - Dariusz Kotlega
- Department of Neurology, Pomeranian Medical University Szczecin, 70-204 Szczecin, Poland;
- Department of Neurology, District Hospital Glogow, 67-200 Glogow, Poland
| | - Ewa Ziemann
- Department of Sport Kinesiology, Poznan University of Physical Education, 61-871 Poznan, Poland;
| |
Collapse
|
66
|
Fleming JW, Capel AJ, Rimington RP, Wheeler P, Leonard AN, Bishop NC, Davies OG, Lewis MP. Bioengineered human skeletal muscle capable of functional regeneration. BMC Biol 2020; 18:145. [PMID: 33081771 PMCID: PMC7576716 DOI: 10.1186/s12915-020-00884-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/30/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Skeletal muscle (SkM) regenerates following injury, replacing damaged tissue with high fidelity. However, in serious injuries, non-regenerative defects leave patients with loss of function, increased re-injury risk and often chronic pain. Progress in treating these non-regenerative defects has been slow, with advances only occurring where a comprehensive understanding of regeneration has been gained. Tissue engineering has allowed the development of bioengineered models of SkM which regenerate following injury to support research in regenerative physiology. To date, however, no studies have utilised human myogenic precursor cells (hMPCs) to closely mimic functional human regenerative physiology. RESULTS Here we address some of the difficulties associated with cell number and hMPC mitogenicity using magnetic association cell sorting (MACS), for the marker CD56, and media supplementation with fibroblast growth factor 2 (FGF-2) and B-27 supplement. Cell sorting allowed extended expansion of myogenic cells and supplementation was shown to improve myogenesis within engineered tissues and force generation at maturity. In addition, these engineered human SkM regenerated following barium chloride (BaCl2) injury. Following injury, reductions in function (87.5%) and myotube number (33.3%) were observed, followed by a proliferative phase with increased MyoD+ cells and a subsequent recovery of function and myotube number. An expansion of the Pax7+ cell population was observed across recovery suggesting an ability to generate Pax7+ cells within the tissue, similar to the self-renewal of satellite cells seen in vivo. CONCLUSIONS This work outlines an engineered human SkM capable of functional regeneration following injury, built upon an open source system adding to the pre-clinical testing toolbox to improve the understanding of basic regenerative physiology.
Collapse
Affiliation(s)
- J W Fleming
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - A J Capel
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - R P Rimington
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - P Wheeler
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - A N Leonard
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - N C Bishop
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - O G Davies
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK
| | - M P Lewis
- School of Sports, Exercise and Health Sciences, Loughborough University, Loughborough, LE11 3TU, UK.
| |
Collapse
|
67
|
Wang L, Shan T. Factors inducing transdifferentiation of myoblasts into adipocytes. J Cell Physiol 2020; 236:2276-2289. [PMID: 32989814 DOI: 10.1002/jcp.30074] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/08/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Fat infiltration in skeletal muscle is observed in several myopathies, is associated with muscular dysfunction, and is strongly correlated with insulin resistance, diabetes, obesity, and aging. In animal production, skeletal muscle fat (also known as intermuscular and intramuscular fat) is positively related to meat quality including tenderness, flavor, and juiciness. Thus, understanding the cell origin and regulation mechanism of skeletal muscle fat infiltration is important for developing therapies against human myopathies as well as for improving meat quality. Notably, age, sarcopenia, oxidative stress, injury, and regeneration can activate adipogenic differentiation potential in myoblasts and affect fat accumulation in skeletal muscle. In addition, several transcriptional and nutritional factors can directly induce transdifferentiation of myoblasts into adipocytes. In this review, we focused on the recent progress in understanding the muscle-to-adipocyte differentiation and summarized and discussed the genetic, nutritional, and physiological factors that can induce transdifferentiation of myoblasts into adipocytes. Moreover, the regulatory roles and mechanisms of these factors during the transdifferentiation process were also discussed.
Collapse
Affiliation(s)
- Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Laboratory of Feed and Animal Nutrition, Hangzhou, China
| |
Collapse
|
68
|
Kim KH, Qiu J, Kuang S. Isolation, Culture, and Differentiation of Primary Myoblasts Derived from Muscle Satellite Cells. Bio Protoc 2020; 10:e3686. [PMID: 33659356 DOI: 10.21769/bioprotoc.3686] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/27/2020] [Accepted: 06/24/2020] [Indexed: 11/02/2022] Open
Abstract
The skeletal muscle is key for body mobility and motor performance, but aging and diseases often lead to progressive loss of muscle mass due to wasting or degeneration of muscle cells. Muscle satellite cells (MuSCs) represent a population of tissue stem cells residing in the skeletal muscles and are responsible for homeostatic maintenance and regeneration of skeletal muscles. Growth, injury, and degenerative signals activate MuSCs, which then proliferate (proliferating MuSCs are called myoblasts), differentiate and fuse with existing multinuclear muscle cells (myofibers) to mediate muscle growth and repair. Here, we describe a protocol for isolating MuSCs from skeletal muscles of mice for in vitro analysis. In addition, we provide a detailed protocol on how to culture and differentiate primary myoblasts into myotubes and an immunofluorescent staining procedure to characterize the cells. These methods are essential for modeling regenerative myogenesis in vitro to understand the dynamics, function and molecular regulation of MuSCs.
Collapse
Affiliation(s)
- Kun Ho Kim
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| |
Collapse
|
69
|
Le Gall L, Ouandaogo ZG, Anakor E, Connolly O, Butler Browne G, Laine J, Duddy W, Duguez S. Optimized method for extraction of exosomes from human primary muscle cells. Skelet Muscle 2020; 10:20. [PMID: 32641118 PMCID: PMC7341622 DOI: 10.1186/s13395-020-00238-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 06/22/2020] [Indexed: 01/19/2023] Open
Abstract
Skeletal muscle is increasingly considered an endocrine organ secreting myokines and extracellular vesicles (exosomes and microvesicles), which can affect physiological changes with an impact on different pathological conditions, including regenerative processes, aging, and myopathies. Primary human myoblasts are an essential tool to study the muscle vesicle secretome. Since their differentiation in conditioned media does not induce any signs of cell death or cell stress, artefactual effects from those processes are unlikely. However, adult human primary myoblasts senesce in long-term tissue culture, so a major technical challenge is posed by the need to avoid artefactual effects resulting from pre-senescent changes. Since these cells should be studied within a strictly controlled pre-senescent division count (<21 divisions), and yields of myoblasts per muscle biopsy are low, it is difficult or impossible to amplify sufficiently large cell numbers (some 250 × 106 myoblasts) to obtain sufficient conditioned medium for the standard ultracentrifugation approach to exosome isolation. Thus, an optimized strategy to extract and study secretory muscle vesicles is needed. In this study, conditions are optimized for the in vitro cultivation of human myoblasts, and the quality and yield of exosomes extracted using an ultracentrifugation protocol are compared with a modified polymer-based precipitation strategy combined with extra washing steps. Both vesicle extraction methods successfully enriched exosomes, as vesicles were positive for CD63, CD82, CD81, floated at identical density (1.15-1.27 g.ml−1), and exhibited similar size and cup-shape using electron microscopy and NanoSight tracking. However, the modified polymer-based precipitation was a more efficient strategy to extract exosomes, allowing their extraction in sufficient quantities to explore their content or to isolate a specific subpopulation, while requiring >30 times fewer differentiated myoblasts than what is required for the ultracentrifugation method. In addition, exosomes could still be integrated into recipient cells such as human myotubes or iPSC-derived motor neurons. Modified polymer-based precipitation combined with extra washing steps optimizes exosome yield from a lower number of differentiated myoblasts and less conditioned medium, avoiding senescence and allowing the execution of multiple experiments without exhausting the proliferative capacity of the myoblasts.
Collapse
Affiliation(s)
- Laura Le Gall
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry~Londonderry, UK
| | | | - Ekene Anakor
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry~Londonderry, UK
| | - Owen Connolly
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry~Londonderry, UK
| | | | - Jeanne Laine
- Centre for Research in Myology, INSERM UMRS_974, Sorbonne Université, Paris, France
| | - William Duddy
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry~Londonderry, UK
| | - Stephanie Duguez
- Northern Ireland Center for Stratified/Personalised Medicine, Biomedical Sciences Research Institute, Ulster University, Derry~Londonderry, UK.
| |
Collapse
|
70
|
Regulation of the Mammalian SWI/SNF Family of Chromatin Remodeling Enzymes by Phosphorylation during Myogenesis. BIOLOGY 2020; 9:biology9070152. [PMID: 32635263 PMCID: PMC7407365 DOI: 10.3390/biology9070152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 11/16/2022]
Abstract
Myogenesis is the biological process by which skeletal muscle tissue forms. Regulation of myogenesis involves a variety of conventional, epigenetic, and epigenomic mechanisms that control chromatin remodeling, DNA methylation, histone modification, and activation of transcription factors. Chromatin remodeling enzymes utilize ATP hydrolysis to alter nucleosome structure and/or positioning. The mammalian SWItch/Sucrose Non-Fermentable (mSWI/SNF) family of chromatin remodeling enzymes is essential for myogenesis. Here we review diverse and novel mechanisms of regulation of mSWI/SNF enzymes by kinases and phosphatases. The integration of classic signaling pathways with chromatin remodeling enzyme function impacts myoblast viability and proliferation as well as differentiation. Regulated processes include the assembly of the mSWI/SNF enzyme complex, choice of subunits to be incorporated into the complex, and sub-nuclear localization of enzyme subunits. Together these processes influence the chromatin remodeling and gene expression events that control myoblast function and the induction of tissue-specific genes during differentiation.
Collapse
|
71
|
Intermittent Hypoxic Exposure with High Dose of Arginine Impact on Circulating Mediators of Tissue Regeneration. Nutrients 2020; 12:nu12071933. [PMID: 32610647 PMCID: PMC7400083 DOI: 10.3390/nu12071933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 12/16/2022] Open
Abstract
Intermittent exposure to hypoxia (IHE) increases production of reactive oxygen and nitrogen species which, as signalling molecules, participate in tissue injury–repair–regeneration cascade. The process is also stimulated by arginine whose bioavailability is a limiting factor for NO synthesis. The effects of IHE in combination with arginine (Arg) intake on myogenesis and angiogenesis mediators were examined in a randomized and placebo-controlled trial. Blood samples were collected from 38 elite athletes on the 1st, 7th and 14th days during the training camp. The oral doses of arginine (2 × 6 g/day) and/or IHE using hypoxicator GO2Altitude (IHE and Arg/IHE) were applied. Serum NO and H2O2 concentrations increased significantly and were related to muscle damage (CK activity >900 IU/mL) in IHE and Arg/IHE compared to placebo. The changes in NO and H2O2 elevated the levels of circulating growth factors such as HGF, IHG-1, PDGFBB, BDNF, VEGF and EPO. Modification of the lipid profile, especially reduced non-HDL, was an additional beneficial effect of hypoxic exposure with arginine intake. Intermittent hypoxic exposure combined with high-dose arginine intake was demonstrated to affect circulating mediators of injury–repair–regeneration. Therefore, a combination of IHE and arginine seems to be a potential therapeutic and non-pharmacological method to modulate the myogenesis and angiogenesis in elite athletes.
Collapse
|
72
|
Farhang-Sardroodi S, Wilkie KP. Mathematical Model of Muscle Wasting in Cancer Cachexia. J Clin Med 2020; 9:jcm9072029. [PMID: 32605273 PMCID: PMC7409297 DOI: 10.3390/jcm9072029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cachexia is a debilitating condition characterized by an extreme loss of skeletal muscle mass, which negatively impacts patients' quality of life, reduces their ability to sustain anti-cancer therapies, and increases the risk of mortality. Recent discoveries have identified the myostatin/activin A/ActRIIB pathway as critical to muscle wasting by inducing satellite cell quiescence and increasing muscle-specific ubiquitin ligases responsible for atrophy. Remarkably, pharmacological blockade of the ActRIIB pathway has been shown to reverse muscle wasting and prolong the survival time of tumor-bearing animals. To explore the implications of this signaling pathway and potential therapeutic targets in cachexia, we construct a novel mathematical model of muscle tissue subjected to tumor-derived cachectic factors. The model formulation tracks the intercellular interactions between cancer cell, satellite cell, and muscle cell populations. The model is parameterized by fitting to colon-26 mouse model data, and the analysis provides insight into tissue growth in healthy, cancerous, and post-cachexia treatment conditions. Model predictions suggest that cachexia fundamentally alters muscle tissue health, as measured by the stem cell ratio, and this is only partially recovered by anti-cachexia treatment. Our mathematical findings suggest that after blocking the myostatin/activin A pathway, partial recovery of cancer-induced muscle loss requires the activation and proliferation of the satellite cell compartment with a functional differentiation program.
Collapse
|
73
|
Moyle LA, Cheng RY, Liu H, Davoudi S, Ferreira SA, Nissar AA, Sun Y, Gentleman E, Simmons CA, Gilbert PM. Three-dimensional niche stiffness synergizes with Wnt7a to modulate the extent of satellite cell symmetric self-renewal divisions. Mol Biol Cell 2020; 31:1703-1713. [PMID: 32491970 PMCID: PMC7521850 DOI: 10.1091/mbc.e20-01-0078] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Satellite cells (SCs), the resident adult stem cells of skeletal muscle, are required for tissue repair throughout life. While many signaling pathways are known to control SC self-renewal, less is known about the mechanisms underlying the spatiotemporal control of self-renewal during skeletal muscle repair. Here, we measured biomechanical changes that accompany skeletal muscle regeneration and determined the implications on SC fate. Using atomic force microscopy, we quantified a 2.9-fold stiffening of the SC niche at time-points associated with planar-oriented symmetric self-renewal divisions. Immunohistochemical analysis confirms increased extracellular matrix deposition within the basal lamina. To test whether three-dimensional (3D) niche stiffness can alter SC behavior or fate, we embedded isolated SC-associated muscle fibers within biochemically inert agarose gels tuned to mimic native tissue stiffness. Time-lapse microscopy revealed that a stiff 3D niche significantly increased the proportion of planar-oriented divisions, without effecting SC viability, fibronectin deposition, or fate change. We then found that 3D niche stiffness synergizes with WNT7a, a biomolecule shown to control SC symmetric self-renewal divisions via the noncanonical WNT/planar cell polarity pathway, to modify stem cell pool expansion. Our results provide new insights into the role of 3D niche biomechanics in regulating SC fate choice.
Collapse
Affiliation(s)
- Louise A Moyle
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Richard Y Cheng
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Haijiao Liu
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.,Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Sadegh Davoudi
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Silvia A Ferreira
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, United Kingdom
| | - Aliyah A Nissar
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
| | - Yu Sun
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, United Kingdom
| | - Craig A Simmons
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.,Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| | - Penney M Gilbert
- Institute of Biomedical Engineering, Toronto, ON M5S 3G9, Canada.,Donnelly Centre for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto ON, M5S 1A8, Canada
| |
Collapse
|
74
|
Gudagudi KB, d’Entrèves NP, Woudberg NJ, Steyn PJ, Myburgh KH. In vitro induction of quiescence in isolated primary human myoblasts. Cytotechnology 2020; 72:189-202. [PMID: 31993891 PMCID: PMC7192999 DOI: 10.1007/s10616-019-00365-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/30/2019] [Indexed: 02/06/2023] Open
Abstract
Adult skeletal muscle stem cells, satellite cells, remain in an inactive or quiescent state in vivo under physiological conditions. Progression through the cell cycle, including activation of quiescent cells, is a tightly regulated process. Studies employing in vitro culture of satellite cells, primary human myoblasts (PHMs), necessitate isolation myoblasts from muscle biopsies. Further studies utilizing these cells should endeavour to represent their native in vivo characteristics as closely as possible, also considering variability between individual donors. This study demonstrates the approach of utilizing KnockOut™ Serum Replacement (KOSR)-supplemented culture media as a quiescence-induction media for 10 days in PHMs isolated and expanded from three different donors. Cell cycle analysis demonstrated that treatment resulted in an increase in G1 phase and decreased S phase proportions in all donors (p < 0.005). The proportions of cells in G1 and G2 phases differed in proliferating myoblasts when comparing donors (p < 0.05 to p < 0.005), but following KOSR treatment, the proportion of cells in G1 (p = 0.558), S (p = 0.606) and G2 phases (p = 0.884) were equivalent between donors. When cultured in the quiescence-induction media, expression of CD34 and Myf5 remained constant above > 98% over time from day 0 to day 10. In contrast activation (CD56), proliferation (Ki67) and myogenic marker MyoD decreased, indicated de-differentiation. Induction of quiescence was accompanied in all three clones by fold change in p21 mRNA greater than 3.5 and up to tenfold. After induction of quiescence, differentiation into myotubes was not affected. In conclusion, we describe a method to induce quiescence in PHMs from different donors.
Collapse
Affiliation(s)
- Kirankumar B. Gudagudi
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| | - Niccolò Passerin d’Entrèves
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| | - Nicholas J. Woudberg
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| | - Paul J. Steyn
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
- Department of Human Biology, University of Cape Town, Anzio Road, Observatory, South Africa
| | - Kathryn H. Myburgh
- Department of Physiological Sciences, Stellenbosch University, Matieland, Private Bag X1, Stellenbosch, 7602 South Africa
| |
Collapse
|
75
|
Narayanan N, Jiang C, Wang C, Uzunalli G, Whittern N, Chen D, Jones OG, Kuang S, Deng M. Harnessing Fiber Diameter-Dependent Effects of Myoblasts Toward Biomimetic Scaffold-Based Skeletal Muscle Regeneration. Front Bioeng Biotechnol 2020; 8:203. [PMID: 32266234 PMCID: PMC7105569 DOI: 10.3389/fbioe.2020.00203] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/02/2020] [Indexed: 12/28/2022] Open
Abstract
Regeneration of skeletal muscles is limited in cases of volumetric muscle loss and muscle degenerative diseases. Therefore, there is a critical need for developing strategies that provide cellular and structural support for skeletal muscle regeneration. In the present work, a bioengineered cell niche composed of mechanically competent aligned polyester fiber scaffolds is developed to mimic the oriented muscle fiber microenvironment by electrospinning poly(lactide-co-glycolide) (PLGA) using a custom-designed rotating collector with interspaced parallel blades. Aligned fiber scaffolds with fiber diameters ranging from 335 ± 154 nm to 3013 ± 531 nm are characterized for their bioactivities in supporting growth and differentiation of myoblasts. During in vitro culture, polymeric scaffolds with larger fiber diameter support enhanced alignment, growth, and differentiation of myoblasts associated with phosphorylation of p38 MAPK and upregulated expression of myogenin and myosin heavy chain. In vivo studies using a dystrophin-deficient mdx mouse model show that optimized fiber scaffolds seeded with primary myoblasts result in formation of dystrophin-positive myofibers network in tibialis anterior muscles. Collectively, these experiments provide critical insights on harnessing interactions between muscle cells and engineered fiber matrices to develop effective biomaterials for accelerated muscle regeneration.
Collapse
Affiliation(s)
- Naagarajan Narayanan
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Chunhui Jiang
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Chao Wang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Gözde Uzunalli
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Nicole Whittern
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Da Chen
- Department of Food Sciences, Purdue University, West Lafayette, IN, United States
| | - Owen G. Jones
- Department of Food Sciences, Purdue University, West Lafayette, IN, United States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Meng Deng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
- Department of Materials Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
76
|
Su Y, Yu Y, Liu C, Zhang Y, Liu C, Ge M, Li L, Lan M, Wang T, Li M, Liu F, Xiong L, Wang K, He T, Shi J, Song Y, Zhao Y, Li N, Yu Z, Meng Q. Fate decision of satellite cell differentiation and self-renewal by miR-31-IL34 axis. Cell Death Differ 2020; 27:949-965. [PMID: 31332295 PMCID: PMC7206105 DOI: 10.1038/s41418-019-0390-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
Affiliation(s)
- Yang Su
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Yingying Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Chuncheng Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
- The Institute of Bioengineering and Technology, Inner Mongolia University of Science and Technology, Baotou, 014010, China
| | - Yuying Zhang
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Chang Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Mengxu Ge
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Lei Li
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Miaomiao Lan
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Tongtong Wang
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Min Li
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Fan Liu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Lei Xiong
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Kun Wang
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Ting He
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Jianyun Shi
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Yongli Song
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Yaofeng Zhao
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Ning Li
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Zhengquan Yu
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China
| | - Qingyong Meng
- State Key Laboratories for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China.
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Haidian District, Beijing, 100193, China.
| |
Collapse
|
77
|
Abstract
The transition between proliferating and quiescent states must be carefully regulated to ensure that cells divide to create the cells an organism needs only at the appropriate time and place. Cyclin-dependent kinases (CDKs) are critical for both transitioning cells from one cell cycle state to the next, and for regulating whether cells are proliferating or quiescent. CDKs are regulated by association with cognate cyclins, activating and inhibitory phosphorylation events, and proteins that bind to them and inhibit their activity. The substrates of these kinases, including the retinoblastoma protein, enforce the changes in cell cycle status. Single cell analysis has clarified that competition among factors that activate and inhibit CDK activity leads to the cell's decision to enter the cell cycle, a decision the cell makes before S phase. Signaling pathways that control the activity of CDKs regulate the transition between quiescence and proliferation in stem cells, including stem cells that generate muscle and neurons. © 2020 American Physiological Society. Compr Physiol 10:317-344, 2020.
Collapse
Affiliation(s)
- Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California, USA.,Department of Biological Chemistry, David Geffen School of Medicine, and the Molecular Biology Institute, University of California, Los Angeles, California, USA.,Molecular Biology Institute, University of California, Los Angeles, California, USA
| |
Collapse
|
78
|
Tiburcy M, Markov A, Kraemer LK, Christalla P, Rave‐Fraenk M, Fischer HJ, Reichardt HM, Zimmermann W. Regeneration competent satellite cell niches in rat engineered skeletal muscle. FASEB Bioadv 2019; 1:731-746. [PMID: 32123818 PMCID: PMC6996344 DOI: 10.1096/fba.2019-00013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 02/21/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022] Open
Abstract
Satellite cells reside in defined niches and are activated upon skeletal muscle injury to facilitate regeneration. Mechanistic studies of skeletal muscle regeneration are hampered by the inability to faithfully simulate satellite cell biology in vitro. We sought to overcome this limitation by developing tissue engineered skeletal muscle (ESM) with (1) satellite cell niches and (2) the capacity to regenerate after injury. ESMs contained quiescent Pax7-positive satellite cells in morphologically defined niches. Satellite cells could be activated to repair (i) cardiotoxin and (ii) mechanical crush injuries. Activation of the Wnt-pathway was essential for muscle regeneration. Finally, muscle progenitors from the engineered niche developed de novo ESM in vitro and regenerated skeletal muscle after cardiotoxin-induced injury in vivo. We conclude that ESM with functional progenitor niches reminiscent of the in vivo satellite cell niches can be engineered in vitro. ESM may ultimately be exploited in disease modeling, drug screening, or muscle regeneration.
Collapse
Affiliation(s)
- Malte Tiburcy
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| | - Alex Markov
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| | - Lena K. Kraemer
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| | - Peter Christalla
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
| | - Margret Rave‐Fraenk
- Department of Radiation Therapy and RadiooncologyGeorg‐August University GöttingenGöttingenGermany
| | - Henrike J. Fischer
- Institute for Cellular and Molecular ImmunologyGeorg‐August University GöttingenGöttingenGermany
- Present address:
Institute for ImmunologyMedical FacultyRWTH Aachen UniversityAachenGermany
| | - Holger M. Reichardt
- Institute for Cellular and Molecular ImmunologyGeorg‐August University GöttingenGöttingenGermany
| | - Wolfram‐Hubertus Zimmermann
- Institute of Pharmacology and ToxicologyGeorg‐August University GöttingenGöttingenGermany
- DZHK (German Center for Cardiovascular Research), partner site GöttingenGöttingenGermany
| |
Collapse
|
79
|
A Three-Dimensional Culture Model of Reversibly Quiescent Myogenic Cells. Stem Cells Int 2019; 2019:7548160. [PMID: 31827532 PMCID: PMC6885280 DOI: 10.1155/2019/7548160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/24/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
Satellite cells (SC) are the stem cells of skeletal muscles. They are quiescent in adult animals but resume proliferation to allow muscle hypertrophy or regeneration after injury. The mechanisms balancing quiescence, self-renewal, and differentiation of SC are difficult to analyze in vivo owing to their complexity and in vitro because the staminal character of SC is lost when they are removed from the niche and is not adequately reproduced in the culture models currently available. To overcome these difficulties, we set up a culture model of the myogenic C2C12 cell line in suspension. When C2C12 cells are cultured in suspension, they enter a state of quiescence and form three-dimensional aggregates (myospheres) that produce the extracellular matrix and express markers of quiescent SC. In the initial phase of culture, a portion of the cells fuses in syncytia and abandons the myospheres. The remaining cells are mononucleated and quiescent but resume proliferation and differentiation when plated in a monolayer. The notch pathway controls the quiescent state of the cells as shown by the fact that its inhibition leads to the resumption of differentiation. Within this context, notch3 appears to play a central role in the activity of this pathway since the expression of notch1 declines soon after aggregation. In summary, the culture model of C2C12 in suspension may be used to study the cellular interactions of muscle stem cells and the pathways controlling SC quiescence entrance and maintenance.
Collapse
|
80
|
Kann AP, Krauss RS. Multiplexed RNAscope and immunofluorescence on whole-mount skeletal myofibers and their associated stem cells. Development 2019; 146:dev179259. [PMID: 31519691 PMCID: PMC6826044 DOI: 10.1242/dev.179259] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022]
Abstract
Skeletal muscle myofibers are large syncytial cells comprising hundreds of myonuclei, and in situ hybridization experiments have reported a range of transcript localization patterns within them. Although some transcripts are uniformly distributed throughout myofibers, proximity to specialized regions can affect the programming of myonuclei and functional compartmentalization of transcripts. Established techniques are limited by a lack of both sensitivity and spatial resolution, restricting the ability to identify different patterns of gene expression. In this study, we adapted RNAscope fluorescent in situ hybridization technology for use on whole-mount mouse primary myofibers, a preparation that isolates single myofibers with their associated muscle stem cells remaining in their niche. This method can be combined with immunofluorescence, enabling an unparalleled ability to visualize and quantify transcripts and proteins across the length and depth of skeletal myofibers and their associated stem cells. Using this approach, we demonstrate a range of potential uses, including the visualization of specialized transcriptional programming within myofibers, tracking activation-induced transcriptional changes, quantification of stem cell heterogeneity and evaluation of stem cell niche factor transcription patterns.
Collapse
Affiliation(s)
- Allison P Kann
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Robert S Krauss
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
81
|
Harvey A, Caretti G, Moresi V, Renzini A, Adamo S. Interplay between Metabolites and the Epigenome in Regulating Embryonic and Adult Stem Cell Potency and Maintenance. Stem Cell Reports 2019; 13:573-589. [PMID: 31597110 PMCID: PMC6830055 DOI: 10.1016/j.stemcr.2019.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/29/2019] [Accepted: 09/06/2019] [Indexed: 12/15/2022] Open
Abstract
The environment surrounding stem cells has the ability to elicit profound, heritable epigenetic changes orchestrated by multiple epigenetic mechanisms, which can be modulated by the level of specific metabolites. In this review, we highlight the significance of metabolism in regulating stem cell homeostasis, cell state, and differentiation capacity, using metabolic regulation of embryonic and adult muscle stem cells as examples, and cast light on the interaction between cellular metabolism and epigenetics. These new regulatory networks, based on the dynamic interplay between metabolism and epigenetics in stem cell biology, are important, not only for understanding tissue homeostasis, but to determine in vitro culture conditions which accurately support normal cell physiology.
Collapse
Affiliation(s)
- Alexandra Harvey
- School of BioSciences, University of Melbourne, Parkville, VIC 2010, Australia
| | - Giuseppina Caretti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Viviana Moresi
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy.
| | - Alessandra Renzini
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy
| | - Sergio Adamo
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy
| |
Collapse
|
82
|
Kargl CK, Nie Y, Evans S, Stout J, Shannahan JH, Kuang S, Gavin TP. Factors secreted from high glucose treated endothelial cells impair expansion and differentiation of human skeletal muscle satellite cells. J Physiol 2019; 597:5109-5124. [DOI: 10.1113/jp278165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
| | - Yaohui Nie
- Department of Health and KinesiologyPurdue University
| | | | | | | | - Shihuan Kuang
- Department of Animal SciencesPurdue University West Lafayette IN USA
| | | |
Collapse
|
83
|
Wang J, Khodabukus A, Rao L, Vandusen K, Abutaleb N, Bursac N. Engineered skeletal muscles for disease modeling and drug discovery. Biomaterials 2019; 221:119416. [PMID: 31419653 DOI: 10.1016/j.biomaterials.2019.119416] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 01/04/2023]
Abstract
Skeletal muscle is the largest organ of human body with several important roles in everyday movement and metabolic homeostasis. The limited ability of small animal models of muscle disease to accurately predict drug efficacy and toxicity in humans has prompted the development in vitro models of human skeletal muscle that fatefully recapitulate cell and tissue level functions and drug responses. We first review methods for development of three-dimensional engineered muscle tissues and organ-on-a-chip microphysiological systems and discuss their potential utility in drug discovery research and development of new regenerative therapies. Furthermore, we describe strategies to increase the functional maturation of engineered muscle, and motivate the importance of incorporating multiple tissue types on the same chip to model organ cross-talk and generate more predictive drug development platforms. Finally, we review the ability of available in vitro systems to model diseases such as type II diabetes, Duchenne muscular dystrophy, Pompe disease, and dysferlinopathy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Lingjun Rao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Keith Vandusen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
84
|
Jia Z, Nie Y, Yue F, Kong Y, Gu L, Gavin TP, Liu X, Kuang S. A requirement of Polo-like kinase 1 in murine embryonic myogenesis and adult muscle regeneration. eLife 2019; 8:e47097. [PMID: 31393265 PMCID: PMC6687435 DOI: 10.7554/elife.47097] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/17/2019] [Indexed: 01/22/2023] Open
Abstract
Muscle development and regeneration require delicate cell cycle regulation of embryonic myoblasts and adult muscle satellite cells (MuSCs). Through analysis of the Polo-like kinase (Plk) family cell-cycle regulators in mice, we show that Plk1's expression closely mirrors myoblast dynamics during embryonic and postnatal myogenesis. Cell-specific deletion of Plk1 in embryonic myoblasts leads to depletion of myoblasts, developmental failure and prenatal lethality. Postnatal deletion of Plk1 in MuSCs does not perturb their quiescence but depletes activated MuSCs as they enter the cell cycle, leading to regenerative failure. The Plk1-null MuSCs are arrested at the M-phase, accumulate DNA damage, and apoptose. Mechanistically, Plk1 deletion upregulates p53, and inhibition of p53 promotes survival of the Plk1-null myoblasts. Pharmacological inhibition of Plk1 similarly inhibits proliferation but promotes differentiation of myoblasts in vitro, and blocks muscle regeneration in vivo. These results reveal for the first time an indispensable role of Plk1 in developmental and regenerative myogenesis.
Collapse
Affiliation(s)
- Zhihao Jia
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Yaohui Nie
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
- Department of Health and KinesiologyPurdue UniversityWest LafayetteUnited States
| | - Feng Yue
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Yifan Kong
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Lijie Gu
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
| | - Timothy P Gavin
- Department of Health and KinesiologyPurdue UniversityWest LafayetteUnited States
| | - Xiaoqi Liu
- Department of BiochemistryPurdue UniversityWest LafayetteUnited States
- Center for Cancer ResearchPurdue UniversityWest LafayetteUnited States
| | - Shihuan Kuang
- Department of Animal SciencesPurdue UniversityWest LafayetteUnited States
- Center for Cancer ResearchPurdue UniversityWest LafayetteUnited States
| |
Collapse
|
85
|
Dunn A, Talovic M, Patel K, Patel A, Marcinczyk M, Garg K. Biomaterial and stem cell-based strategies for skeletal muscle regeneration. J Orthop Res 2019; 37:1246-1262. [PMID: 30604468 DOI: 10.1002/jor.24212] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 12/13/2018] [Indexed: 02/04/2023]
Abstract
Adult skeletal muscle can regenerate effectively after mild physical or chemical insult. Muscle trauma or disease can overwhelm this innate capacity for regeneration and result in heightened inflammation and fibrotic tissue deposition resulting in loss of structure and function. Recent studies have focused on biomaterial and stem cell-based therapies to promote skeletal muscle regeneration following injury and disease. Many stem cell populations besides satellite cells are implicated in muscle regeneration. These stem cells include but are not limited to mesenchymal stem cells, adipose-derived stem cells, hematopoietic stem cells, pericytes, fibroadipogenic progenitors, side population cells, and CD133+ stem cells. However, several challenges associated with their isolation, availability, delivery, survival, engraftment, and differentiation have been reported in recent studies. While acellular scaffolds offer a relatively safe and potentially off-the-shelf solution to cell-based therapies, they are often unable to stimulate host cell migration and activity to a level that would result in clinically meaningful regeneration of traumatized muscle. Combining stem cells and biomaterials may offer a viable therapeutic strategy that may overcome the limitations associated with these therapies when they are used in isolation. In this article, we review the stem cell populations that can stimulate muscle regeneration in vitro and in vivo. We also discuss the regenerative potential of combination therapies that utilize both stem cell and biomaterials for the treatment of skeletal muscle injury and disease. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1246-1262, 2019.
Collapse
Affiliation(s)
- Andrew Dunn
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Muhamed Talovic
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Krishna Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Anjali Patel
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Madison Marcinczyk
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, Saint Louis, Missouri
| |
Collapse
|
86
|
Kikani CK, Wu X, Fogarty S, Kang SAW, Dephoure N, Gygi SP, Sabatini DM, Rutter J. Activation of PASK by mTORC1 is required for the onset of the terminal differentiation program. Proc Natl Acad Sci U S A 2019; 116:10382-10391. [PMID: 31072927 PMCID: PMC6534978 DOI: 10.1073/pnas.1804013116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
During skeletal muscle regeneration, muscle stem cells (MuSCs) respond to multiple signaling inputs that converge onto mammalian target of rapamycin complex 1 (mTORC1) signaling pathways. mTOR function is essential for establishment of the differentiation-committed progenitors (early stage of differentiation, marked by the induction of myogenin expression), myotube fusion, and, ultimately, hypertrophy (later stage of differentiation). While a major mTORC1 substrate, p70S6K, is required for myotube fusion and hypertrophy, an mTORC1 effector for the induction of myogenin expression remains unclear. Here, we identified Per-Arnt-Sim domain kinase (PASK) as a downstream phosphorylation target of mTORC1 in MuSCs during differentiation. We have recently shown that the PASK phosphorylates Wdr5 to stimulate MuSC differentiation by epigenetically activating the myogenin promoter. We show that phosphorylation of PASK by mTORC1 is required for the activation of myogenin transcription, exit from self-renewal, and induction of the myogenesis program. Our studies reveal that mTORC1-PASK signaling is required for the rise of myogenin-positive committed myoblasts (early stage of myogenesis), whereas mTORC1-S6K signaling is required for myoblast fusion (later stage of myogenesis). Thus, our discoveries allow molecular dissection of mTOR functions during different stages of the myogenesis program driven by two different substrates.
Collapse
Affiliation(s)
- Chintan K Kikani
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84132;
| | - Xiaoying Wu
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Sarah Fogarty
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84132
| | - Seong Anthony Woo Kang
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Noah Dephoure
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142
- Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84132;
- Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84132
| |
Collapse
|
87
|
Pardo-Saganta A, Calvo IA, Saez B, Prosper F. Role of the Extracellular Matrix in Stem Cell Maintenance. CURRENT STEM CELL REPORTS 2019. [DOI: 10.1007/s40778-019-0149-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
88
|
Song JY, Pineault KM, Wellik DM. Development, repair, and regeneration of the limb musculoskeletal system. Curr Top Dev Biol 2019; 132:451-486. [PMID: 30797517 DOI: 10.1016/bs.ctdb.2018.12.011] [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] [Indexed: 12/14/2022]
Abstract
The limb musculoskeletal system provides a primary means for locomotion, manipulation of objects and protection for most vertebrate organisms. Intricate integration of the bone, tendon and muscle tissues are required for function. These three tissues arise largely independent of one another, but the connections formed during later development are maintained throughout life and are re-established following injury. Each of these tissues also have mesenchymal stem/progenitor cells that function in maintenance and repair. Here in, we will review the major events in the development of limb skeleton, tendon, and muscle tissues, their response to injury, and discuss current knowledge regarding resident progenitor/stem cells within each tissue that participate in development, repair, and regeneration in vivo.
Collapse
Affiliation(s)
- Jane Y Song
- Program in Cell and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
| | - Kyriel M Pineault
- Department of Cell & Regenerative Biology, University of Wisconsin, Madison, WI, United States
| | - Deneen M Wellik
- Department of Cell & Regenerative Biology, University of Wisconsin, Madison, WI, United States.
| |
Collapse
|
89
|
Singh A, Yadav CB, Tabassum N, Bajpeyee AK, Verma V. Stem cell niche: Dynamic neighbor of stem cells. Eur J Cell Biol 2018; 98:65-73. [PMID: 30563738 DOI: 10.1016/j.ejcb.2018.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/09/2018] [Accepted: 12/11/2018] [Indexed: 12/19/2022] Open
Abstract
Stem cell niche is a specialized and dynamic microenvironment around the stem cells which plays a critical role in maintaining the stemness properties of stem cells. Over the years, advancement in the research activity has revealed the various important aspects of stem cell niche including cell-cell interaction, cell-extracellular matrix interaction, a large number of soluble signaling factors and various biochemical and biophysical cues (such as oxygen tension, flow, and shear and pore size). Stem cells have the potential to be a powerful tool in regenerative medicine due to their self-renewal property and immense differentiation potential. Recent progresses in in vitro culture conditions of embryonic stem cells, adult stem cells and induced pluripotent stem cells have enabled the researchers to investigate and understand the role of the microenvironment in stem cell properties. The engineered artificial stem cell niche has led to a better execution of stem cells in regenerative medicine. Here we elucidate the key components of stem cell niche and their role in niche engineering and stem cell therapeutics.
Collapse
Affiliation(s)
- Anshuman Singh
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - C B Yadav
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - N Tabassum
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - A K Bajpeyee
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - V Verma
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India.
| |
Collapse
|
90
|
Weledji EP, Eyongeta D, Ngounou E. The anatomy of urination: What every physician should know. Clin Anat 2018; 32:60-67. [PMID: 30303589 DOI: 10.1002/ca.23296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/15/2018] [Accepted: 10/03/2018] [Indexed: 12/22/2022]
Abstract
Normal voiding needs a coordinated, sustained bladder contraction of adequate size and duration. It requires a decrease in resistance of the bladder neck and urethra and no obstruction. Voiding problems can arise from abnormal storage of urine or problems with urinary control. The aim of this article was to review the functional anatomy and physiology of urinary control and micturition and the pathophysiology of urinary control problems. The Medline (PubMed) database, Cochrane Library, and Science Citation Index were searched electronically to identify original published studies on bladder anatomy, function and urinary control. References were searched from relevant chapters in specialized texts and all were included. Voiding problems are the most common presenting urological symptoms in general medical practice. Urinary incontinence occurs when the normal process of storing and passing urine is disrupted. A history of coexisting fecal incontinence suggests a neuropathic etiology. A better understanding of the physiology of urinary control could lead to preventive measures for postoperative urinary retention and incontinence such as fluid restriction and to appropriate anesthesia/analgesia, autonomic nerve preservation, total mesorectal excision (TME) for rectal cancer, and biofeedback exercises. It could also suggest appropriate therapeutic measures for established urinary incontinence. Clin. Anat., 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Elroy P Weledji
- Departments of Anatomy and Surgery, Faculty of Health Sciences, University of Buea, Gastrointestinal Surgeon, Regional Hospital Limbe, Limbe, Cameroon
| | - Divine Eyongeta
- Departments of Anatomy and Surgery, Faculty of Health Sciences, University of Buea, Urologist, Regional Hospital, Limbe, Cameroon
| | - Eleanor Ngounou
- Department of Anatomy, Faculty of Health Sciences, University of Buea, Limbe, Cameroon
| |
Collapse
|
91
|
Augmentation of Dermal Wound Healing by Adipose Tissue-Derived Stromal Cells (ASC). Bioengineering (Basel) 2018; 5:bioengineering5040091. [PMID: 30373121 PMCID: PMC6316823 DOI: 10.3390/bioengineering5040091] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/21/2018] [Accepted: 10/24/2018] [Indexed: 12/14/2022] Open
Abstract
The skin is the largest organ of the human body and is the first line of defense against physical and biological damage. Thus, the skin is equipped to self-repair and regenerates after trauma. Skin regeneration after damage comprises a tightly spatial-temporally regulated process of wound healing that involves virtually all cell types in the skin. Wound healing features five partially overlapping stages: homeostasis, inflammation, proliferation, re-epithelization, and finally resolution or fibrosis. Dysreguled wound healing may resolve in dermal scarring. Adipose tissue is long known for its suppressive influence on dermal scarring. Cultured adipose tissue-derived stromal cells (ASCs) secrete a plethora of regenerative growth factors and immune mediators that influence processes during wound healing e.g., angiogenesis, modulation of inflammation and extracellular matrix remodeling. In clinical practice, ASCs are usually administered as part of fractionated adipose tissue i.e., as part of enzymatically isolated SVF (cellular SVF), mechanically isolated SVF (tissue SVF), or as lipograft. Enzymatic isolation of SVF obtained adipose tissue results in suspension of adipocyte-free cells (cSVF) that lack intact intercellular adhesions or connections to extracellular matrix (ECM). Mechanical isolation of SVF from adipose tissue destructs the parenchyma (adipocytes), which results in a tissue SVF (tSVF) with intact connections between cells, as well as matrix. To date, due to a lack of well-designed prospective randomized clinical trials, neither cSVF, tSVF, whole adipose tissue, or cultured ASCs can be indicated as the preferred preparation procedure prior to therapeutic administration. In this review, we present and discuss current literature regarding the different administration options to apply ASCs (i.e., cultured ASCs, cSVF, tSVF, and lipografting) to augment dermal wound healing, as well as the available indications for clinical efficacy.
Collapse
|
92
|
Correra RM, Ollitrault D, Valente M, Mazzola A, Adalsteinsson BT, Ferguson-Smith AC, Marazzi G, Sassoon DA. The imprinted gene Pw1/Peg3 regulates skeletal muscle growth, satellite cell metabolic state, and self-renewal. Sci Rep 2018; 8:14649. [PMID: 30279563 PMCID: PMC6168517 DOI: 10.1038/s41598-018-32941-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/18/2018] [Indexed: 12/16/2022] Open
Abstract
Pw1/Peg3 is an imprinted gene expressed from the paternally inherited allele. Several imprinted genes, including Pw1/Peg3, have been shown to regulate overall body size and play a role in adult stem cells. Pw1/Peg3 is expressed in muscle stem cells (satellite cells) as well as a progenitor subset of muscle interstitial cells (PICs) in adult skeletal muscle. We therefore examined the impact of loss-of-function of Pw1/Peg3 during skeletal muscle growth and in muscle stem cell behavior. We found that constitutive loss of Pw1/Peg3 function leads to a reduced muscle mass and myofiber number. In newborn mice, the reduction in fiber number is increased in homozygous mutants as compared to the deletion of only the paternal Pw1/Peg3 allele, indicating that the maternal allele is developmentally functional. Constitutive and a satellite cell-specific deletion of Pw1/Peg3, revealed impaired muscle regeneration and a reduced capacity of satellite cells for self-renewal. RNA sequencing analyses revealed a deregulation of genes that control mitochondrial function. Consistent with these observations, Pw1/Peg3 mutant satellite cells displayed increased mitochondrial activity coupled with accelerated proliferation and differentiation. Our data show that Pw1/Peg3 regulates muscle fiber number determination during fetal development in a gene-dosage manner and regulates satellite cell metabolism in the adult.
Collapse
Affiliation(s)
- Rosa Maria Correra
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
| | - David Ollitrault
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Mariana Valente
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France
| | - Alessia Mazzola
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France
| | - Bjorn T Adalsteinsson
- Department of Physiology Development and Neuroscience, Downing Street, University of Cambridge, Cambridge, United Kingdom
| | - Anne C Ferguson-Smith
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, United Kingdom
| | - Giovanna Marazzi
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France.
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France.
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France.
| | - David A Sassoon
- UMR S 1166 INSERM (Stem Cells and Regenerative Medicine Team), University of Pierre and Marie Curie Paris VI, Paris, 75634, France.
- Institute of Cardiometabolism and Nutrition (ICAN), Paris, 75013, France.
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Université René Descartes Paris, Paris, France.
| |
Collapse
|
93
|
Brown JL, Lee DE, Rosa-Caldwell ME, Brown LA, Perry RA, Haynie WS, Huseman K, Sataranatarajan K, Van Remmen H, Washington TA, Wiggs MP, Greene NP. Protein imbalance in the development of skeletal muscle wasting in tumour-bearing mice. J Cachexia Sarcopenia Muscle 2018; 9:987-1002. [PMID: 30328290 PMCID: PMC6204589 DOI: 10.1002/jcsm.12354] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 08/28/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cancer cachexia occurs in approximately 80% of cancer patients and is a key contributor to cancer-related death. The mechanisms controlling development of tumour-induced muscle wasting are not fully elucidated. Specifically, the progression and development of cancer cachexia are underexplored. Therefore, we examined skeletal muscle protein turnover throughout the development of cancer cachexia in tumour-bearing mice. METHODS Lewis lung carcinoma (LLC) was injected into the hind flank of C57BL6/J mice at 8 weeks age with tumour allowed to develop for 1, 2, 3, or 4 weeks and compared with PBS injected control. Muscle size was measured by cross-sectional area analysis of haematoxylin and eosin stained tibialis anterior muscle. 2 H2 O was used to assess protein synthesis throughout the development of cancer cachexia. Immunoblot and RT-qPCR were used to measure regulators of protein turnover. TUNEL staining was utilized to measure apoptotic nuclei. LLC conditioned media (LCM) treatment of C2C12 myotubes was used to analyse cancer cachexia in vitro. RESULTS Muscle cross-sectional area decreased ~40% 4 weeks following tumour implantation. Myogenic signalling was suppressed in tumour-bearing mice as soon as 1 week following tumour implantation, including lower mRNA contents of Pax7, MyoD, CyclinD1, and Myogenin, when compared with control animals. AchRδ and AchRε mRNA contents were down-regulated by ~50% 3 weeks following tumour implantation. Mixed fractional synthesis rate protein synthesis was ~40% lower in 4 week tumour-bearing mice when compared with PBS controls. Protein ubiquitination was elevated by ~50% 4 weeks after tumour implantation. Moreover, there was an increase in autophagy machinery after 4 weeks of tumour growth. Finally, ERK and p38 MAPK phosphorylations were fourfold and threefold greater than control muscle 4 weeks following tumour implantation, respectively. Inhibition of p38 MAPK, but not ERK MAPK, in vitro partially rescued LCM-induced loss of myotube diameter. CONCLUSIONS Our findings work towards understanding the pathophysiological signalling in skeletal muscle in the initial development of cancer cachexia. Shortly following the onset of the tumour-bearing state alterations in myogenic regulatory factors are apparent, suggesting early onset alterations in the capacity for myogenic induction. Cancer cachexia presents with a combination of a loss of protein synthesis and increased markers of protein breakdown, specifically in the ubiquitin-proteasome system. Also, p38 MAPK may be a potential therapeutic target to combat cancer cachexia via a p38-FOX01-atrogene-ubiquitin-proteasome mechanism.
Collapse
Affiliation(s)
- Jacob L Brown
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - David E Lee
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Megan E Rosa-Caldwell
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Lemuel A Brown
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA.,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Richard A Perry
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Wesley S Haynie
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Kendra Huseman
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK, 73104, USA
| | - Kavithalakshmi Sataranatarajan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK, 73104, USA
| | - Holly Van Remmen
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 N.E. 13th Street, Oklahoma City, OK, 73104, USA.,Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Michael P Wiggs
- Integrated Physiology and Nutrition Laboratory, Department of Health and Kinesiology, University of Texas at Tyler, Tyler, TX, 75799, USA
| | - Nicholas P Greene
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, AR, 72701, USA
| |
Collapse
|
94
|
Virgilio KM, Martin KS, Peirce SM, Blemker SS. Agent-based model illustrates the role of the microenvironment in regeneration in healthy and mdx skeletal muscle. J Appl Physiol (1985) 2018; 125:1424-1439. [PMID: 30070607 DOI: 10.1152/japplphysiol.00379.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease with no effective treatment. Multiple mechanisms are thought to contribute to muscle wasting, including increased susceptibility to contraction-induced damage, chronic inflammation, fibrosis, altered satellite stem cell (SSC) dynamics, and impaired regenerative capacity. The goals of this project were to 1) develop an agent-based model of skeletal muscle that predicts the dynamic regenerative response of muscle cells, fibroblasts, SSCs, and inflammatory cells as a result of contraction-induced injury, 2) calibrate and validate the model parameters based on comparisons with published experimental measurements, and 3) use the model to investigate how changing isolated and combined factors known to be associated with DMD (e.g., altered fibroblast or SSC behaviors) influence muscle regeneration. Our predictions revealed that the percent of injured muscle that recovered 28 days after injury was dependent on the peak SSC counts following injury. In simulations with near-full cross-sectional area recovery (healthy, 4-wk mdx, 3-mo mdx), the SSC counts correlated with the extent of initial injury; however, in simulations with impaired regeneration (9-mo mdx), the peak SSC counts were suppressed relative to initial injury. The differences in SSC counts between these groups were emergent predictions dependent on altered microenvironment factors known to be associated with DMD. Multiple cell types influenced the peak number of SSCs, but no individual parameter predicted the differences in SSC counts. This finding suggests that interventions to target the microenvironment rather than SSCs directly could be an effective method for improving regeneration in impaired muscle. NEW & NOTEWORTHY A computational model predicted that satellite stem cell (SSC) counts are correlated with muscle cross-sectional area (CSA) recovery following injury. In simulations with impaired CSA recovery, SSC counts are suppressed relative to healthy muscle. The suppressed SSC counts were an emergent model prediction, because all simulations had equal initial SSC counts. Fibroblast and anti-inflammatory macrophage counts influenced SSC counts, but no single factor was able to predict the pathological differences in SSC counts that lead to impaired regeneration.
Collapse
Affiliation(s)
- Kelley M Virgilio
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
| | - Kyle S Martin
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
| | - Silvia S Blemker
- Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia.,Department of Orthopaedic Surgery, University of Virginia , Charlottesville, Virginia.,Department of Mechanical and Aerospace Engineering, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
95
|
Joanisse S, Snijders T, Nederveen JP, Parise G. The Impact of Aerobic Exercise on the Muscle Stem Cell Response. Exerc Sport Sci Rev 2018; 46:180-187. [DOI: 10.1249/jes.0000000000000153] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
96
|
Biomaterials in Tendon and Skeletal Muscle Tissue Engineering: Current Trends and Challenges. MATERIALS 2018; 11:ma11071116. [PMID: 29966303 PMCID: PMC6073924 DOI: 10.3390/ma11071116] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/20/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022]
Abstract
Tissue engineering is a promising approach to repair tendon and muscle when natural healing fails. Biohybrid constructs obtained after cells’ seeding and culture in dedicated scaffolds have indeed been considered as relevant tools for mimicking native tissue, leading to a better integration in vivo. They can also be employed to perform advanced in vitro studies to model the cell differentiation or regeneration processes. In this review, we report and analyze the different solutions proposed in literature, for the reconstruction of tendon, muscle, and the myotendinous junction. They classically rely on the three pillars of tissue engineering, i.e., cells, biomaterials and environment (both chemical and physical stimuli). We have chosen to present biomimetic or bioinspired strategies based on understanding of the native tissue structure/functions/properties of the tissue of interest. For each tissue, we sorted the relevant publications according to an increasing degree of complexity in the materials’ shape or manufacture. We present their biological and mechanical performances, observed in vitro and in vivo when available. Although there is no consensus for a gold standard technique to reconstruct these musculo-skeletal tissues, the reader can find different ways to progress in the field and to understand the recent history in the choice of materials, from collagen to polymer-based matrices.
Collapse
|
97
|
Huang Y, Lai X, Hu L, Lei C, Lan X, Zhang C, Ma Y, Zheng L, Bai Y, Lin F, Chen H. Over‐expression of DEC1 inhibits myogenic differentiation by modulating MyoG activity in bovine satellite cell. J Cell Physiol 2018; 233:9365-9374. [DOI: 10.1002/jcp.26471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/05/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Yongzhen Huang
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| | - Xinsheng Lai
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
- The Laboratory of Synaptic Development and Plasticity, Institute of Life ScienceNanchang UniversityNanchangChina
- School of Life ScienceNanchang UniversityNanchangChina
| | - Linyong Hu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau BiologyChinese Academy of SciencesXiningQinghaiChina
| | - Chuzhao Lei
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| | - Xianyong Lan
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| | - Chunlei Zhang
- Institute of Cellular and Molecular BiologyJiangsu Normal UniversityXuzhouJiangsuChina
| | - Yun Ma
- College of Life Sciences, Xinyang Normal UniversityInstitute for Conservation and Utilization of Agro‐Bioresources in Dabie MountainsXinyangHenanChina
| | - Li Zheng
- Henan University of Animal Husbandry and EconomyZhengzhouHenanChina
| | - Yue‐Yu Bai
- Animal Health Supervision in Henan ProvinceZhengzhouHenanChina
| | - Fengpeng Lin
- Bureau of Animal Husbandry of Biyang CountyBiyangHenanChina
| | - Hong Chen
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| |
Collapse
|
98
|
Preclinical characterization of the JAK/STAT inhibitor SGI-1252 on skeletal muscle function, morphology, and satellite cell content. PLoS One 2018; 13:e0198611. [PMID: 29897957 PMCID: PMC5999283 DOI: 10.1371/journal.pone.0198611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/22/2018] [Indexed: 01/20/2023] Open
Abstract
Background Recent studies have highlighted the JAK/STAT signaling pathway in the regulation of muscle satellite cell behavior. Herein we report preclinical studies designed to characterize the effects of a novel JAK/STAT inhibitor on plantar flexor skeletal muscle function, morphology, and satellite cell content. Methods The compound, SGI-1252, was administered orally (400mg/kg) in a 10% dextrose solution to wild type mice (n = 6) 3 times per week for 8 weeks. A control group (n = 6) received only the dextrose solution. Results SGI-1252 was well tolerated, as animals displayed similar weight gain over the 8-week treatment period. Following treatment, fatigue in the gastrocnemius-soleus-plantaris complex was greater in the SGI-1252 mice during a 300 second tetanic contraction bout (p = 0.035), though both the rate of fatigue and maximal force production were similar. SGI-1252 treated mice had increased type II myofiber cross-sectional area (1434.8 ± 225.4 vs 1754.7 ± 138.5 μm2), along with an increase in wet muscle mass (125.45 ± 5.46 vs 139.6 ± 12.34 mg, p = 0.032) of the gastrocnemius relative to vehicle treated mice. SGI-1252 treatment reduced gastrocnemius STAT3 phosphorylation 53% (94.79 ± 45.9 vs 44.5 ± 6.1 MFI) and significantly increased the concentration of Pax7+ satellite cells (2589.2 ± 105.5 vs 2859.4 ± 177.5 SC/mm3) in the gastrocnemius. SGI-1252 treatment suppressed MyoD (p = 0.013) and Myogenin (p<0.0001) expression in human primary myoblasts, resulting in reduced myogenic differentiation (p = 0.039). Conclusions Orally delivered SGI-1252 was well tolerated, attenuates skeletal muscle STAT3 activity, and increases satellite cell content in mouse gastrocnemius muscle, likely by inhibiting myogenic progression.
Collapse
|
99
|
Alonso-Martin S, Auradé F, Mademtzoglou D, Rochat A, Zammit PS, Relaix F. SOXF factors regulate murine satellite cell self-renewal and function through inhibition of β-catenin activity. eLife 2018; 7:26039. [PMID: 29882512 PMCID: PMC6021169 DOI: 10.7554/elife.26039] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/07/2018] [Indexed: 12/17/2022] Open
Abstract
Muscle satellite cells are the primary source of stem cells for postnatal skeletal muscle growth and regeneration. Understanding genetic control of satellite cell formation, maintenance, and acquisition of their stem cell properties is on-going, and we have identified SOXF (SOX7, SOX17, SOX18) transcriptional factors as being induced during satellite cell specification. We demonstrate that SOXF factors regulate satellite cell quiescence, self-renewal and differentiation. Moreover, ablation of Sox17 in the muscle lineage impairs postnatal muscle growth and regeneration. We further determine that activities of SOX7, SOX17 and SOX18 overlap during muscle regeneration, with SOXF transcriptional activity requisite. Finally, we show that SOXF factors also control satellite cell expansion and renewal by directly inhibiting the output of β-catenin activity, including inhibition of Ccnd1 and Axin2. Together, our findings identify a key regulatory function of SoxF genes in muscle stem cells via direct transcriptional control and interaction with canonical Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Sonia Alonso-Martin
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Frédéric Auradé
- Sorbonne Université, INSERM U974, Center for Research in Myology, Paris, France
| | - Despoina Mademtzoglou
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France
| | - Anne Rochat
- Sorbonne Université, INSERM U974, Center for Research in Myology, Paris, France
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Frédéric Relaix
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10, Créteil, France.,Université Paris Est, Faculté de Medecine, Créteil, France.,Ecole Nationale Veterinaire d'Alfort, Maison Alfort, France.,Etablissement Français du Sang, Creteil, France.,APHP, Hopitaux UniversitairesHenri Mondor, Centre de Référence des Maladies Neuromusculaires GNMH, Créteil, France
| |
Collapse
|
100
|
Regeneration of diaphragm with bio-3D cellular patch. Biomaterials 2018; 167:1-14. [DOI: 10.1016/j.biomaterials.2018.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/22/2022]
|