1
|
Fu X, Zhang F, Dong X, Pu L, Feng Y, Xu Y, Gao F, Liang T, Kang J, Sun H, Hong T, Liu Y, Zhou H, Jiang J, Yin D, Hu X, Wang DZ, Ding J, Chen J. Adapting cytoskeleton-mitochondria patterning with myocyte differentiation by promyogenic PRR33. Cell Death Differ 2024:10.1038/s41418-024-01363-w. [PMID: 39147882 DOI: 10.1038/s41418-024-01363-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/02/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Coordinated cytoskeleton-mitochondria organization during myogenesis is crucial for muscle development and function. Our understanding of the underlying regulatory mechanisms remains inadequate. Here, we identified a novel muscle-enriched protein, PRR33, which is upregulated during myogenesis and acts as a promyogenic factor. Depletion of Prr33 in C2C12 represses myoblast differentiation. Genetic deletion of Prr33 in mice reduces myofiber size and decreases muscle strength. The Prr33 mutant mice also exhibit impaired myogenesis and defects in muscle regeneration in response to injury. Interactome and transcriptome analyses reveal that PRR33 regulates cytoskeleton and mitochondrial function. Remarkably, PRR33 interacts with DESMIN, a key regulator of cytoskeleton-mitochondria organization in muscle cells. Abrogation of PRR33 in myocytes substantially abolishes the interaction of DESMIN filaments with mitochondria, leading to abnormal intracellular accumulation of DESMIN and mitochondrial disorganization/dysfunction in myofibers. Together, our findings demonstrate that PRR33 and DESMIN constitute an important regulatory module coordinating mitochondrial organization with muscle differentiation.
Collapse
Affiliation(s)
- Xuyang Fu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Feng Zhang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Xiaoxuan Dong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Linbin Pu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Yan Feng
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yang Xu
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Feng Gao
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tian Liang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Jianmeng Kang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Hongke Sun
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Tingting Hong
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yunxia Liu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hongmei Zhou
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jun Jiang
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Deling Yin
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xinyang Hu
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Da-Zhi Wang
- University of South Florida Health Heart Institute, Center for Regenerative Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33602, USA
| | - Jian Ding
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Jinghai Chen
- Department of Cardiology of Second Affiliated Hospital, State Key Laboratory of Transvascular Implantation Devices, Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| |
Collapse
|
2
|
Serna C, Sandepudi K, Keate RL, Zhang SL, Cotton KY, De La Isla A, Murillo M, Bouricha Y, Domenighetti AA, Franz CK, Jordan SW. Incorporation of decellularized-ECM in graphene-based scaffolds enhances axonal outgrowth and branching in neuro-muscular co-cultures. Sci Prog 2024; 107:368504241281469. [PMID: 39314156 PMCID: PMC11423365 DOI: 10.1177/00368504241281469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Peripheral nerve and large-scale muscle injuries result in significant disability, necessitating the development of biomaterials that can restore functional deficits by promoting tissue regrowth in an electroactive environment. Among these materials, graphene is favored for its high conductivity, but its low bioactivity requires enhancement through biomimetic components. In this study, we extrusion printed graphene-poly(lactide-co-glycolide) (graphene) lattice scaffolds, aiming to increase bioactivity by incorporating decellularized extracellular matrix (dECM) derived from mouse pup skeletal muscle. We first evaluated these scaffolds using human-induced pluripotent stem cell (hiPSC)-derived motor neurons co-cultured with supportive glia, observing significant improvements in axon outgrowth. Next, we tested the scaffolds with C2C12 mouse and human primary myoblasts, finding no significant differences in myotube formation between dECM-graphene and graphene scaffolds. Finally, using a more complex hiPSC-derived 3D motor neuron spheroid model co-cultured with human myoblasts, we demonstrated that dECM-graphene scaffolds significantly improved axonal expansion towards peripheral myoblasts and increased axonal network density compared to graphene-only scaffolds. Features of early neuromuscular junction formation were identified near neuromuscular interfaces in both scaffold types. These findings suggest that dECM-graphene scaffolds are promising candidates for enhancing neuromuscular regeneration, offering robust support for the growth and development of diverse neuromuscular tissues.
Collapse
Affiliation(s)
- Carlos Serna
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Kirtana Sandepudi
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Rebecca L Keate
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, USA
| | - Sophia L Zhang
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Kristen Y Cotton
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Alberto De La Isla
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| | - Matias Murillo
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Yasmine Bouricha
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Andrea A Domenighetti
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Colin K Franz
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
- Physical Medicine and Rehabilitation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ken & Ruth Dave Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Kimberly K. Querrey and Louis A. Simpson Institute for Bioelectronics, Simpson Querrey Biomedical Research Center, Chicago, IL, USA
| | - Sumanas W Jordan
- Division of Plastic Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Simpson Querrey Institute, Northwestern University, Chicago, IL, USA
- Biologics Laboratory, Shirley Ryan AbilityLab, Chicago, IL, USA
| |
Collapse
|
3
|
Barutcu AR, Elizalde G, Gonzalez AE, Soni K, Rinn JL, Wagers AJ, Almada AE. Prolonged FOS activity disrupts a global myogenic transcriptional program by altering 3D chromatin architecture in primary muscle progenitor cells. Skelet Muscle 2022; 12:20. [PMID: 35971133 PMCID: PMC9377060 DOI: 10.1186/s13395-022-00303-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/04/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The AP-1 transcription factor, FBJ osteosarcoma oncogene (FOS), is induced in adult muscle satellite cells (SCs) within hours following muscle damage and is required for effective stem cell activation and muscle repair. However, why FOS is rapidly downregulated before SCs enter cell cycle as progenitor cells (i.e., transiently expressed) remains unclear. Further, whether boosting FOS levels in the proliferating progeny of SCs can enhance their myogenic properties needs further evaluation. METHODS We established an inducible, FOS expression system to evaluate the impact of persistent FOS activity in muscle progenitor cells ex vivo. We performed various assays to measure cellular proliferation and differentiation, as well as uncover changes in RNA levels and three-dimensional (3D) chromatin interactions. RESULTS Persistent FOS activity in primary muscle progenitor cells severely antagonizes their ability to differentiate and form myotubes within the first 2 weeks in culture. RNA-seq analysis revealed that ectopic FOS activity in muscle progenitor cells suppressed a global pro-myogenic transcriptional program, while activating a stress-induced, mitogen-activated protein kinase (MAPK) transcriptional signature. Additionally, we observed various FOS-dependent, chromosomal re-organization events in A/B compartments, topologically associated domains (TADs), and genomic loops near FOS-regulated genes. CONCLUSIONS Our results suggest that elevated FOS activity in recently activated muscle progenitor cells perturbs cellular differentiation by altering the 3D chromosome organization near critical pro-myogenic genes. This work highlights the crucial importance of tightly controlling FOS expression in the muscle lineage and suggests that in states of chronic stress or disease, persistent FOS activity in muscle precursor cells may disrupt the muscle-forming process.
Collapse
Affiliation(s)
- A Rasim Barutcu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Present address: Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Gabriel Elizalde
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alfredo E Gonzalez
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Kartik Soni
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - John L Rinn
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Present address: BioFrontiers and Department of Biochemistry, University of Colorado Boulder, Boulder, CO, 80303, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Albert E Almada
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Department of Orthopaedic Surgery, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
4
|
KYMASIN UP Natural Product Inhibits Osteoclastogenesis and Improves Osteoblast Activity by Modulating Src and p38 MAPK. Nutrients 2022; 14:nu14153053. [PMID: 35893905 PMCID: PMC9370798 DOI: 10.3390/nu14153053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
The imbalance in osteoblast (OB)-dependent bone formation in favor of osteoclast (OC)-dependent bone resorption is the main cause of loss of tissue mineral mass during bone remodeling leading to osteoporosis conditions. Thus, the suppression of OC activity together with the improvement in the OB activity has been proposed as an effective therapy for maintaining bone mass during aging. We tested the new dietary product, KYMASIN UP containing standardized Withania somnifera, Silybum marianum and Trigonella foenum-graecum herbal extracts or the single extracts in in vitro models mimicking osteoclastogenesis (i.e., RAW 264.7 cells treated with RANKL, receptor activator of nuclear factor kappa-Β ligand) and OB differentiation (i.e., C2C12 myoblasts treated with BMP2, bone morphogenetic protein 2). We found that the dietary product reduces RANKL-dependent TRAP (tartrate-resistant acid phosphatase)-positive cells (i.e., OCs) formation and TRAP activity, and down-regulates osteoclastogenic markers by reducing Src (non-receptor tyrosine kinase) and p38 MAPK (mitogen-activated protein kinase) activation. Withania somnifera appears as the main extract responsible for the anti-osteoclastogenic effect of the product. Moreover, KYMASIN UP maintains a physiological release of the soluble decoy receptor for RANKL, OPG (osteoprotegerin), in osteoporotic conditions and increases calcium mineralization in C2C12-derived OBs. Interestingly, KYMASIN UP induces differentiation in human primary OB-like cells derived from osteoporotic subjects. Based on our results, KYMASIN UP or Withania somnifera-based dietary supplements might be suggested to reverse the age-related functional decline of bone tissue by re-balancing the activity of OBs and OCs, thus improving the quality of life in the elderly and reducing social and health-care costs.
Collapse
|
5
|
Desmin deficiency affects the microenvironment of the cardiac side population and Sca1+ stem cell population of the adult heart and impairs their cardiomyogenic commitment. Cell Tissue Res 2022; 389:309-326. [DOI: 10.1007/s00441-022-03643-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/20/2022] [Indexed: 11/02/2022]
|
6
|
Kihara Y, Homma J, Takagi R, Ishigaki K, Nagata S, Yamato M. Laminin-221-derived recombinant fragment facilitates isolation of cultured skeletal myoblasts. Regen Ther 2022; 20:147-156. [PMID: 35620637 PMCID: PMC9111930 DOI: 10.1016/j.reth.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/30/2022] [Accepted: 04/26/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Laminin is a major component of the basement membrane, containing multiple domains that bind integrin, collagen, nidogen, dystroglycan, and heparan sulfate. Laminin-221, expressed in skeletal and cardiac muscles, has strong affinity for the cell-surface receptor, integrin α7X2β1. The E8 domain of laminin-221, which is essential for cell integrin binding, is commercially available as a purified recombinant protein fragment. In this study, recombinant E8 fragment was used to purify primary rodent myoblasts. We established a facile and inexpensive method for primary myoblast culture exploiting the high affinity binding of integrin α7X2β1 to laminin-221. Methods Total cell populations from dissociated muscle tissue were enzymatically digested and seeded onto laminin-221 E8 fragment-coated dishes. The culture medium containing non-adherent floating cells was removed after 2-hour culture at 37 °C. The adherent cells were subjected to immunofluorescence staining of desmin, differentiation experiments, and gene expression analysis. Results The cells obtained were 70.3 ± 5.49% (n = 5) desmin positive in mouse and 67.7 ± 1.65% (n = 3) in rat. Immunofluorescent staining and gene expression analyses of cultured cells showed phenotypic traits of myoblasts. Conclusion This study reports a novel facile method for primary culture of myoblasts obtained from mouse and rat skeletal muscle by exploiting the high affinity of integrin α7X2β1 to laminin-221. Myoblasts are muscle progenitor cells that differentiate into skeletal muscle. Various methods have been reported to isolate myoblasts, such as FACS and MACS. Integrin α7X2, predominantly expressed in myocytes and cardiomyocytes, binds laminin-221 with high affinity. We established a novel method for primary culture of myoblasts by utilizing the high affinity of integrin α7X2β1 to laminin-221.
Collapse
Affiliation(s)
- Yuki Kihara
- Department of Pediatrics, Tokyo Women's Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Jun Homma
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Ryo Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Keiko Ishigaki
- Department of Pediatrics, Tokyo Women's Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Satoru Nagata
- Department of Pediatrics, Tokyo Women's Medical University, School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
- Corresponding author. Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan. Tel: +81 3-3353-8111, Fax: +81 3-3359-6046
| |
Collapse
|
7
|
Direct Conversion of Bovine Dermal Fibroblasts into Myotubes by Viral Delivery of Transcription Factor bMyoD. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Direct reprogramming of somatic cells to myoblasts and myotubes holds great potential for muscle development, disease modeling and regenerative medicine. According to recent studies, direct conversion of fibroblasts to myoblasts was performed by using a transcription factor, myoblast determination protein (MyoD), which belongs to a family of myogenic regulatory factors. Therefore, MyoD is considered to be a key driver in the generation of induced myoblasts. In this study, we compared the direct phenotypic conversion of bovine dermal fibroblasts (BDFs) into myoblasts and myotubes by supplementing a transcription factor, bovine MyoD (bMyoD), in the form of recombinant protein or the bMyoD gene, through retroviral vectors. As a result, the delivery of the bMyoD gene to BDFs was more efficient for inducing reprogramming, resulting in direct conversion to myoblasts and myotubes, when compared with protein delivery. BDFs cultured with retrovirus encoding bMyoD increased myogenic gene expression, such as MyoG, MYH3 and MYMK. In addition, the cells expressed myoblast or myotube-specific marker proteins, MyoG and Desmin, respectively. Our findings provide an informative tool for the myogenesis of domestic-animal-derived somatic cells via transgenic technology. By using this method, a new era of regenerative medicine and cultured meat is expected.
Collapse
|
8
|
Sugimoto T, Imai S, Yoshikawa M, Fujisato T, Hashimoto T, Nakamura T. Mechanical unloading in 3D-engineered muscle leads to muscle atrophy by suppressing protein synthesis. J Appl Physiol (1985) 2022; 132:1091-1103. [PMID: 35297688 DOI: 10.1152/japplphysiol.00323.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Three dimensional (3D)-engineered muscle is an useful approach to a more comprehensive understanding of molecular mechanisms underlying unloading-induced muscle atrophy. We investigated the effects of mechanical unloading on molecular muscle protein synthesis (MPS)- and muscle protein breakdown (MPB)-related signaling pathways involved in muscle atrophy in 3D-engineered muscle, and to better understand in vitro model of muscle disuse. The 3D-engineered muscle consisting of C2C12 myoblasts and type-1 collagen gel was allowed to differentiate for 2 weeks and divided into three groups: 0 days of stretched-on control (CON), 2 and/or 7 days of stretched-on (ON), in which both ends of the muscle were fixed with artificial tendons, and the stretched-off group (OFF), in which one side of the artificial tendon was detached. Muscle weight (-38.1 to -48.4%), length (-67.0 to -73.5%), twitch contractile force (-70.5 to -75.0%) and myosin heavy chain expression (-32.5 to -50.5%) in the OFF group were significantly decreased on days 2 and 7 compared with the ON group (P < 0.05, respectively), despite that ON group was stable over time. Although determinative molecular signaling could not be identified, the MPS rate reflected by puromysin labeled protein was significantly decreased following mechanical unloading (P < 0.05, -38.5 to -51.1%). Meanwhile, MPB, particularly the ubiquitin-proteasome pathway, was not impacted. Hence, mechanical unloading of 3D-engineered muscle in vitro leads to muscle atrophy by suppressing MPS, cell differentiation, and cell growth rather than the promotion of MPB.
Collapse
Affiliation(s)
- Takeshi Sugimoto
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Shoma Imai
- Division of Human Sciences, Faculty of Engineering, Osaka Institute of Technology, Ohmiya, Osaka, Japan
| | - Maki Yoshikawa
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Toshia Fujisato
- Biomedical Engineering Graduate School of Engineering, Osaka Institute of Technology, Ohmiya, Osaka, Japan
| | - Takeshi Hashimoto
- Faculty of Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Tomohiro Nakamura
- Division of Human Sciences, Faculty of Engineering, Osaka Institute of Technology, Ohmiya, Osaka, Japan
| |
Collapse
|
9
|
Kural Mangit E, Boustanabadimaralan Düz N, Dinçer P. A cytoplasmic escapee: desmin is going nuclear. Turk J Biol 2022; 45:711-719. [PMID: 35068951 PMCID: PMC8733954 DOI: 10.3906/biy-2107-54] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/04/2021] [Indexed: 11/03/2022] Open
Abstract
It has been a long time since researchers have focused on the cytoskeletal proteins' unconventional functions in the nucleus. Subcellular localization of a protein not only affects its functions but also determines the accessibility for cellular processes. Desmin is a muscle-specific, cytoplasmic intermediate filament protein, the cytoplasmic roles of which are defined. Yet, there is some evidence pointing out nuclear functions for desmin. In silico and wet lab analysis shows that desmin can enter and function in the nucleus. Furthermore, the candidate nuclear partners of desmin support the notion that desmin can serve as a transcriptional regulator inside the nucleus. Uncovering the nuclear functions and partners of desmin will provide a new insight into the biological significance of desmin.
Collapse
Affiliation(s)
- Ecem Kural Mangit
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara Turkey.,Laboratory Animals Research and Application Centre, Hacettepe University, Ankara Turkey
| | | | - Pervin Dinçer
- Department of Medical Biology, Faculty of Medicine, Hacettepe University, Ankara Turkey
| |
Collapse
|
10
|
Major E, Keller I, Horváth D, Tamás I, Erdődi F, Lontay B. Smoothelin-Like Protein 1 Regulates the Thyroid Hormone-Induced Homeostasis and Remodeling of C2C12 Cells via the Modulation of Myosin Phosphatase. Int J Mol Sci 2021; 22:10293. [PMID: 34638630 PMCID: PMC8508602 DOI: 10.3390/ijms221910293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 11/28/2022] Open
Abstract
The pathological elevation of the active thyroid hormone (T3) level results in the manifestation of hyperthyroidism, which is associated with alterations in the differentiation and contractile function of skeletal muscle (SKM). Myosin phosphatase (MP) is a major cellular regulator that hydrolyzes the phosphoserine of phosphorylated myosin II light chain. MP consists of an MYPT1/2 regulatory and a protein phosphatase 1 catalytic subunit. Smoothelin-like protein 1 (SMTNL1) is known to inhibit MP by directly binding to MP as well as by suppressing the expression of MYPT1 at the transcriptional level. Supraphysiological vs. physiological concentration of T3 were applied on C2C12 myoblasts and differentiated myotubes in combination with the overexpression of SMTNL1 to assess the role and regulation of MP under these conditions. In non-differentiated myoblasts, MP included MYPT1 in the holoenzyme complex and its expression and activity was regulated by SMTNL1, affecting the phosphorylation level of MLC20 assessed using semi-quantitative Western blot analysis. SMTNL1 negatively influenced the migration and cytoskeletal remodeling of myoblasts measured by high content screening. In contrast, in myotubes, the expression of MYPT2 but not MYPT1 increased in a T3-dependent and SMTNL1-independent manner. T3 treatment combined with SMTNL1 overexpression impeded the activity of MP. In addition, MP interacted with Na+/K+-ATPase and dephosphorylated its inhibitory phosphorylation sites, identifying this protein as a novel MP substrate. These findings may help us gain a better understanding of myopathy, muscle weakness and the disorder of muscle regeneration in hyperthyroid patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Beáta Lontay
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (E.M.); (I.K.); (D.H.); (I.T.); (F.E.)
| |
Collapse
|
11
|
Nin DS, Wujanto C, Tan TZ, Lim D, Damen JMA, Wu KY, Dai ZM, Lee ZW, Idres SB, Leong YH, Jha S, Ng JSY, Low JJH, Chang SC, Tan DSP, Wu W, Choo BA, Deng LW. GAGE mediates radio resistance in cervical cancers via the regulation of chromatin accessibility. Cell Rep 2021; 36:109621. [PMID: 34469741 DOI: 10.1016/j.celrep.2021.109621] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 06/03/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022] Open
Abstract
Radiotherapy (RT) resistance is a major cause of treatment failure in cancers that use definitive RT as their primary treatment modality. This study identifies the cancer/testis (CT) antigen G antigen (GAGE) as a mediator of radio resistance in cervical cancers. Elevated GAGE expression positively associates with de novo RT resistance in clinical samples. GAGE, specifically the GAGE12 protein variant, confers RT resistance through synemin-dependent chromatin localization, promoting the association of histone deacetylase 1/2 (HDAC1/2) to its inhibitor actin. This cumulates to elevated histone 3 lysine 56 acetylation (H3K56Ac) levels, increased chromatin accessibility, and improved DNA repair efficiency. Molecular or pharmacological disruption of the GAGE-associated complex restores radiosensitivity. Molecularly, this study demonstrates the role of GAGE in the regulation of chromatin dynamics. Clinically, this study puts forward the utility of GAGE as a pre-screening biomarker to identify poor responders at initial diagnosis and the therapeutic potential of agents that target GAGE and its associated complex in combination with radiotherapy to improve outcomes.
Collapse
Affiliation(s)
- Dawn Sijin Nin
- Department of Biochemistry, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore (NUS), Singapore 117596, Singapore; NUS Center for Cancer Research, YLLSOM, NUS, Singapore 117599, Singapore.
| | - Caryn Wujanto
- Department of Radiation Oncology, National University Hospital (NUH), Singapore 119074, Singapore; National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, NUS, Singapore 117599, Singapore
| | - Diana Lim
- Department of Pathology, NUH, Singapore 119074, Singapore; National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore
| | - J Mirjam A Damen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht , the Netherlands
| | - Kuan-Yi Wu
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ziyu Melvin Dai
- Department of Biochemistry, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore (NUS), Singapore 117596, Singapore
| | - Zheng-Wei Lee
- Department of Biochemistry, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore (NUS), Singapore 117596, Singapore
| | - Shabana Binte Idres
- Department of Biochemistry, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore (NUS), Singapore 117596, Singapore
| | - Yiat Horng Leong
- Department of Radiation Oncology, National University Hospital (NUH), Singapore 119074, Singapore; National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore
| | - Sudhakar Jha
- Department of Biochemistry, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore (NUS), Singapore 117596, Singapore; Cancer Science Institute of Singapore, NUS, Singapore 117599, Singapore; National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore; Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA; NUS Center for Cancer Research, YLLSOM, NUS, Singapore 117599, Singapore
| | - Joseph Soon-Yau Ng
- National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore; Department of Obstetrics and Gynecology, YLLSOM, NUS, Singapore 119228, Singapore
| | - Jeffrey J H Low
- National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore; Department of Obstetrics and Gynecology, YLLSOM, NUS, Singapore 119228, Singapore
| | - Shih-Chung Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - David Shao Peng Tan
- Cancer Science Institute of Singapore, NUS, Singapore 117599, Singapore; National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore; Department of Hematology-Oncology, NUHS, Singapore 119228, Singapore; NUS Center for Cancer Research, YLLSOM, NUS, Singapore 117599, Singapore
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Padualaan 8, 3584 CH Utrecht , the Netherlands
| | - Bok Ai Choo
- Department of Radiation Oncology, National University Hospital (NUH), Singapore 119074, Singapore; National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore
| | - Lih-Wen Deng
- Department of Biochemistry, Yong Loo Lin School of Medicine (YLLSOM), National University of Singapore (NUS), Singapore 117596, Singapore; National University Cancer Institute, Singapore National University Health System (NUHS), Singapore 119074, Singapore; NUS Center for Cancer Research, YLLSOM, NUS, Singapore 117599, Singapore; NUS Graduate School - Integrative Sciences and Engineering Programme, NUS, Singapore 119077, Singapore.
| |
Collapse
|
12
|
Skeletal and Cardiac Muscle Disorders Caused by Mutations in Genes Encoding Intermediate Filament Proteins. Int J Mol Sci 2021; 22:ijms22084256. [PMID: 33923914 PMCID: PMC8073371 DOI: 10.3390/ijms22084256] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023] Open
Abstract
Intermediate filaments are major components of the cytoskeleton. Desmin and synemin, cytoplasmic intermediate filament proteins and A-type lamins, nuclear intermediate filament proteins, play key roles in skeletal and cardiac muscle. Desmin, encoded by the DES gene (OMIM *125660) and A-type lamins by the LMNA gene (OMIM *150330), have been involved in striated muscle disorders. Diseases include desmin-related myopathy and cardiomyopathy (desminopathy), which can be manifested with dilated, restrictive, hypertrophic, arrhythmogenic, or even left ventricular non-compaction cardiomyopathy, Emery–Dreifuss Muscular Dystrophy (EDMD2 and EDMD3, due to LMNA mutations), LMNA-related congenital Muscular Dystrophy (L-CMD) and LMNA-linked dilated cardiomyopathy with conduction system defects (CMD1A). Recently, mutations in synemin (SYNM gene, OMIM *606087) have been linked to cardiomyopathy. This review will summarize clinical and molecular aspects of desmin-, lamin- and synemin-related striated muscle disorders with focus on LMNA and DES-associated clinical entities and will suggest pathogenetic hypotheses based on the interplay of desmin and lamin A/C. In healthy muscle, such interplay is responsible for the involvement of this network in mechanosignaling, nuclear positioning and mitochondrial homeostasis, while in disease it is disturbed, leading to myocyte death and activation of inflammation and the associated secretome alterations.
Collapse
|
13
|
Kural-Mangıt E, Dinçer PR. Physical evidence on desmin-lamin B interaction. Cytoskeleton (Hoboken) 2021; 78:14-17. [PMID: 33475247 DOI: 10.1002/cm.21651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/31/2020] [Accepted: 01/14/2021] [Indexed: 01/22/2023]
Abstract
Desmin is a muscle specific intermediate filament protein located in cytoplasm. Lamin B, on the other hand, is a nuclear intermediate filament protein. There are studies suggesting a possible interaction between desmin and lamin B yet there is no physical evidence. In the present study, we have shown for the first time a physical interaction between desmin and lamin B via reciprocal co-immunoprecipitation from muscle tissue of wild type AB zebrafish (Danio rerio, Hamilton). The interaction between desmin and lamin B might be a lead on a novel nucleocytoplasmic communication network.
Collapse
Affiliation(s)
- Ecem Kural-Mangıt
- Faculty of Medicine, Department of Medical Biology, Hacettepe University, Ankara, Turkey.,Laboratory Animals Research and Application Center, Hacettepe University, Ankara, Turkey
| | - Pervin Rukiye Dinçer
- Faculty of Medicine, Department of Medical Biology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
14
|
Cohen S. Role of calpains in promoting desmin filaments depolymerization and muscle atrophy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118788. [DOI: 10.1016/j.bbamcr.2020.118788] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022]
|
15
|
Shen T, Lin J, Li X, Deng D. Intermediate filaments in the medial rectus muscles in patients with concomitant exotropia. Int Ophthalmol 2019; 40:403-410. [PMID: 31630292 DOI: 10.1007/s10792-019-01197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/10/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE Distribution of intermediate filament (IF) proteins in normal extraocular muscles (EOMs) showed that the EOMs differ significantly from the other muscles in the body with respect to their IFs composition, including desmin and nestin. The aim of the present study was to investigate the pathological changes in the medial rectus (MR) in patients with concomitant exotropia (XT). METHODS Forty-six MR muscle samples from 46 patients with XT were analyzed pathologically and processed for immunohistochemistry with specific antibodies against desmin and nestin. RESULTS Although most of MR muscles remained normal structures relatively, they presented high expression of desmin, and in contrast, nestin was absent in a large proportion of the MR muscles. CONCLUSION Desmin, which is downregulated in normal EOMs, had high expression in MR muscles of patients with XT. Nestin, which is present in a high proportion of normal EOMs, was downregulated in MR muscles of patients with XT.
Collapse
Affiliation(s)
- Tao Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jing Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xiuling Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Daming Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
16
|
Lieber RL. Biomechanical response of skeletal muscle to eccentric contractions. JOURNAL OF SPORT AND HEALTH SCIENCE 2018; 7:294-309. [PMID: 30356666 PMCID: PMC6189273 DOI: 10.1016/j.jshs.2018.06.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/29/2017] [Accepted: 01/09/2018] [Indexed: 05/18/2023]
Abstract
The forced lengthening of an activated skeletal muscle has been termed an eccentric contraction (EC). This review highlights the mechanically unique nature of the EC and focuses on the specific disruption of proteins within the cell known as cytoskeletal proteins. The major intermediate filament cytoskeletal protein, desmin, has been the focus of work in this area because changes to desmin occur within minutes of ECs and because desmin has been shown to play both a mechanical and biologic role in a muscle's response to EC. It is hoped that these types of studies will assist in decreasing the incidence of muscle injury in athletes and facilitating the development of new therapies to treat muscle injuries.
Collapse
|
17
|
Hol EM, Capetanaki Y. Type III Intermediate Filaments Desmin, Glial Fibrillary Acidic Protein (GFAP), Vimentin, and Peripherin. Cold Spring Harb Perspect Biol 2017; 9:9/12/a021642. [PMID: 29196434 DOI: 10.1101/cshperspect.a021642] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SummaryType III intermediate filament (IF) proteins assemble into cytoplasmic homopolymeric and heteropolymeric filaments with other type III and some type IV IFs. These highly dynamic structures form an integral component of the cytoskeleton of muscle, brain, and mesenchymal cells. Here, we review the current ideas on the role of type III IFs in health and disease. It turns out that they not only offer resilience to mechanical strains, but, most importantly, they facilitate very efficiently the integration of cell structure and function, thus providing the necessary scaffolds for optimal cellular responses upon biochemical stresses and protecting against cell death, disease, and aging.
Collapse
Affiliation(s)
- Elly M Hol
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, The Netherlands.,Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands.,Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, 1098 XH Amsterdam, The Netherlands
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| |
Collapse
|
18
|
Kabagambe SK, Lankford L, Kumar P, Chen YJ, Herout KT, Lee CJ, Stark RA, Farmer DL, Wang A. Isolation of myogenic progenitor cell population from human placenta: A pilot study. J Pediatr Surg 2017; 52:2078-2082. [PMID: 28964407 DOI: 10.1016/j.jpedsurg.2017.08.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 08/28/2017] [Indexed: 01/07/2023]
Abstract
PURPOSE The purpose of this study was to demonstrate a method of isolating myogenic progenitor cells from human placenta chorionic villi and to confirm the myogenic characteristics of the isolated cells. METHODS Cells were isolated from chorionic villi of a second trimester male placenta via a combined enzymatic digestion and explant culture. A morphologically distinct subpopulation of elongated and multinucleated cells was identified. This subpopulation was manually passaged from the explant culture, expanded, and analyzed by fluorescence in situ hybridization (FISH) assay, immunocytochemistry, and flow cytometry. Myogenic characteristics including alignment and fusion were tested by growing these cells on aligned polylactic acid microfibrous scaffold in a fusion media composed of 2% horse serum in Dulbecco's modified Eagle medium/high glucose. RESULTS The expanded subpopulation was uniformly positive for integrin α-7. Presence of Y-chromosome by FISH analysis confirmed chorionic villus origin rather than maternal cell contamination. Isolated cells grew, aligned, and fused on the microfibrous scaffold, and they expressed myogenin, desmin, and MHC confirming their myogenic identity. CONCLUSION Myogenic progenitor cells can be isolated from human chorionic villi. This opens the possibility for translational and clinical applications using autologous myogenic cells for possible engraftment in treatment of chest and abdominal wall defects.
Collapse
Affiliation(s)
| | - Lee Lankford
- University of California, Davis Health, Sacramento, CA, USA
| | | | - Y Julia Chen
- University of California, Davis Health, Sacramento, CA, USA
| | - Kyle T Herout
- University of California, Davis Health, Sacramento, CA, USA
| | - Chelsey J Lee
- University of California, Davis Health, Sacramento, CA, USA
| | | | - Diana L Farmer
- University of California, Davis Health, Sacramento, CA, USA
| | - Aijun Wang
- University of California, Davis Health, Sacramento, CA, USA
| |
Collapse
|
19
|
Zhang S, Chen X, Huang Z, Chen D, Yu B, He J, Zheng P, Yu J, Luo J, Luo Y, Chen H. Effects of MicroRNA-27a on Myogenin Expression and Akt/FoxO1 Signal Pathway during Porcine Myoblast Differentiation. Anim Biotechnol 2017; 29:183-189. [DOI: 10.1080/10495398.2017.1348357] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Shurun Zhang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Junqiu Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Hong Chen
- College of Food Science, Sichuan Agricultural University, Yaan, Sichuan, P. R. China
| |
Collapse
|
20
|
Rodriguez J, Pierre N, Naslain D, Bontemps F, Ferreira D, Priem F, Deldicque L, Francaux M. Urolithin B, a newly identified regulator of skeletal muscle mass. J Cachexia Sarcopenia Muscle 2017; 8:583-597. [PMID: 28251839 PMCID: PMC5566634 DOI: 10.1002/jcsm.12190] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 01/01/2017] [Accepted: 01/10/2017] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The control of muscle size is an essential feature of health. Indeed, skeletal muscle atrophy leads to reduced strength, poor quality of life, and metabolic disturbances. Consequently, strategies aiming to attenuate muscle wasting and to promote muscle growth during various (pathological) physiological states like sarcopenia, immobilization, malnutrition, or cachexia are needed to address this extensive health issue. In this study, we tested the effects of urolithin B, an ellagitannin-derived metabolite, on skeletal muscle growth. METHODS C2C12 myotubes were treated with 15 μM of urolithin B for 24 h. For in vivo experiments, mice were implanted with mini-osmotic pumps delivering continuously 10 μg/day of urolithin B during 28 days. Muscle atrophy was studied in mice with a sciatic nerve denervation receiving urolithin B by the same way. RESULTS Our experiments reveal that urolithin B enhances the growth and differentiation of C2C12 myotubes by increasing protein synthesis and repressing the ubiquitin-proteasome pathway. Genetic and pharmacological arguments support an implication of the androgen receptor. Signalling analyses suggest a crosstalk between the androgen receptor and the mTORC1 pathway, possibly via AMPK. In vivo experiments confirm that urolithin B induces muscle hypertrophy in mice and reduces muscle atrophy after the sciatic nerve section. CONCLUSIONS This study highlights the potential usefulness of urolithin B for the treatment of muscle mass loss associated with various (pathological) physiological states.
Collapse
Affiliation(s)
- Julie Rodriguez
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium.,PROCELL nutrition sprl, 2 Rue Jean Burgers, 7850, Enghien, Belgium
| | - Nicolas Pierre
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| | - Damien Naslain
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| | - Françoise Bontemps
- De Duve Institute, Université catholique de Louvain, 75 Avenue Hippocrate, 1200, Brussels, Belgium
| | - Daneel Ferreira
- Department of Biomolecular Sciences, Division of Pharmacognosy, Research Institute of Pharmaceutical Sciences, University of Mississippi, Medicinal Plant Garden, RM 104, University, MS, 38677, USA
| | - Fabian Priem
- PROCELL nutrition sprl, 2 Rue Jean Burgers, 7850, Enghien, Belgium
| | - Louise Deldicque
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| | - Marc Francaux
- Institute of Neuroscience, Université catholique de Louvain, 1 place Pierre de Coubertin, 1348, Louvain-la-Neuve, Belgium
| |
Collapse
|
21
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
22
|
Fuchs C, Gawlas S, Heher P, Nikouli S, Paar H, Ivankovic M, Schultheis M, Klammer J, Gottschamel T, Capetanaki Y, Weitzer G. Desmin enters the nucleus of cardiac stem cells and modulates Nkx2.5 expression by participating in transcription factor complexes that interact with the nkx2.5 gene. Biol Open 2016; 5:140-53. [PMID: 26787680 PMCID: PMC4823984 DOI: 10.1242/bio.014993] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/13/2015] [Indexed: 12/30/2022] Open
Abstract
The transcription factor Nkx2.5 and the intermediate filament protein desmin are simultaneously expressed in cardiac progenitor cells during commitment of primitive mesoderm to the cardiomyogenic lineage. Up-regulation of Nkx2.5 expression by desmin suggests that desmin may contribute to cardiogenic commitment and myocardial differentiation by directly influencing the transcription of the nkx2.5 gene in cardiac progenitor cells. Here, we demonstrate that desmin activates transcription of nkx2.5 reporter genes, rescues nkx2.5 haploinsufficiency in cardiac progenitor cells, and is responsible for the proper expression of Nkx2.5 in adult cardiac side population stem cells. These effects are consistent with the temporary presence of desmin in the nuclei of differentiating cardiac progenitor cells and its physical interaction with transcription factor complexes bound to the enhancer and promoter elements of the nkx2.5 gene. These findings introduce desmin as a newly discovered and unexpected player in the regulatory network guiding cardiomyogenesis in cardiac stem cells.
Collapse
Affiliation(s)
- Christiane Fuchs
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Sonja Gawlas
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Philipp Heher
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Sofia Nikouli
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 115 27, Greece
| | - Hannah Paar
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Mario Ivankovic
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Martina Schultheis
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Julia Klammer
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Teresa Gottschamel
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 115 27, Greece
| | - Georg Weitzer
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna Biocenter, Vienna A1030, Austria
| |
Collapse
|
23
|
Diokmetzidou A, Tsikitis M, Nikouli S, Kloukina I, Tsoupri E, Papathanasiou S, Psarras S, Mavroidis M, Capetanaki Y. Strategies to Study Desmin in Cardiac Muscle and Culture Systems. Methods Enzymol 2015; 568:427-59. [PMID: 26795479 DOI: 10.1016/bs.mie.2015.09.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Intermediate filament (IF) cytoskeleton comprises the fine-tuning cellular machinery regulating critical homeostatic mechanisms. In skeletal and cardiac muscle, deficiency or disturbance of the IF network leads to severe pathology, particularly in the latter. The three-dimensional scaffold of the muscle-specific IF protein desmin interconnects key features of the cardiac muscle cells, including the Z-disks, intercalated disks, plasma membrane, nucleus, mitochondria, lysosomes, and potentially sarcoplasmic reticulum. This is crucial for the highly organized striated muscle, in which effective energy production and transmission as well as mechanochemical signaling are tightly coordinated among the organelles and the contractile apparatus. The role of desmin and desmin-associated proteins in the biogenesis, trafficking, and organelle function, as well as the development, differentiation, and survival of the cardiac muscle begins to be enlightened, but the precise mechanisms remain elusive. We propose a set of experimental tools that can be used, in vivo and in vitro, to unravel crucial new pathways by which the IF cytoskeleton facilitates proper organelle function, homeostasis, and cytoprotection and further understand how its disturbance and deficiency lead to disease.
Collapse
Affiliation(s)
- Antigoni Diokmetzidou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Mary Tsikitis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Sofia Nikouli
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Ismini Kloukina
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Elsa Tsoupri
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Stamatis Papathanasiou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens, Greece.
| |
Collapse
|
24
|
Yang X, Lai P, Chen XG, Liu ZP, Tang YM, Rao S, Wen CJ, Jiao SR, Lei J. Maternal high-fat diet exposure leads to insulin resistance and impacts myogenic and adipogenicgene expression in offspring rats. EUR J LIPID SCI TECH 2015. [DOI: 10.1002/ejlt.201400421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Xiao Yang
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Peng Lai
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Xiang-Gui Chen
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Zhen-Ping Liu
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Yuan-Mou Tang
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Su Rao
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Chun-Juan Wen
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Shi-Rong Jiao
- School of Bioengineering; Xihua University; Chengdu P. R. China
| | - Ji Lei
- School of Bioengineering; Xihua University; Chengdu P. R. China
| |
Collapse
|
25
|
Li Y, Wang Y, Willems E, Willemsen H, Franssens L, Buyse J, Decuypere E, Everaert N. In ovo L-arginine supplementation stimulates myoblast differentiation but negatively affects muscle development of broiler chicken after hatching. J Anim Physiol Anim Nutr (Berl) 2015; 100:167-77. [PMID: 25846259 DOI: 10.1111/jpn.12299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 01/19/2015] [Indexed: 02/01/2023]
Abstract
In this study, we tested the hypothesis that in ovo feeding (IOF) of L-arginine (L-Arg) enhances nitric oxide (NO) production, stimulates the process of myogenesis, and regulates post-hatching muscle growth. Different doses of L-Arg were injected into the amnion of chicken embryos at embryonic day (ED) 16. After hatching, the body weight of individual male chickens was recorded weekly for 3 weeks. During in vitro experiments, myoblasts of the pectoralis major (PM) were extracted at ED16 and were incubated in medium containing 0.01 mm L-Arg, 0.05 mm L-Arg, and (or) 0.05 mm L-nitro-arginine-methyl-ester (L-NAME), an inhibitor of nitric oxide synthase (NOS). When 25 mg/kg L-Arg/initial egg weight was injected, no difference was observed in body weight at hatch, but a significant decrease was found during the following 3 weeks compared to that of the non-injected and saline-injected control, and this also affected the growth of muscle mass. L-NAME inhibited gene expression of myogenic differentiation antigen (MyoD), myogenin, NOS, and follistatin, decreased the cell viability, and increased myostatin (MSTN) gene expression. 0.05 mm L-Arg stimulated myogenin gene expression but also depressed muscle cell viability. L-NAME blocked the effect of 0.05 mm L-Arg on myogenin mRNA levels when co-incubated with 0.05 mm L-Arg. L-Arg treatments had no significant influence on NOS mRNA gene expression, but had inhibiting effect on follistatin gene expression, while L-NAME treatments had effects on both. These results suggested that L-Arg stimulated myoblast differentiation, but the limited number of myoblasts would form less myotubes and then less myofibers, while the latter limited the growth of muscle mass.
Collapse
Affiliation(s)
- Y Li
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium.,Laboratory of Translational Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Y Wang
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium
| | - E Willems
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium
| | - H Willemsen
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium
| | - L Franssens
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium
| | - J Buyse
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium
| | - E Decuypere
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium
| | - N Everaert
- Department of Biosystems, Division of Livestock-Nutrition-Quality, KU Leuven, Leuven, Belgium.,Animal Science Unit, Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| |
Collapse
|
26
|
Harada A, Mallappa C, Okada S, Butler JT, Baker SP, Lawrence JB, Ohkawa Y, Imbalzano AN. Spatial re-organization of myogenic regulatory sequences temporally controls gene expression. Nucleic Acids Res 2015; 43:2008-21. [PMID: 25653159 PMCID: PMC4344497 DOI: 10.1093/nar/gkv046] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/05/2014] [Accepted: 01/13/2015] [Indexed: 12/14/2022] Open
Abstract
During skeletal muscle differentiation, the activation of some tissue-specific genes occurs immediately while others are delayed. The molecular basis controlling temporal gene regulation is poorly understood. We show that the regulatory sequences, but not other regions of genes expressed at late times of myogenesis, are in close physical proximity in differentiating embryonic tissue and in differentiating culture cells, despite these genes being located on different chromosomes. Formation of these inter-chromosomal interactions requires the lineage-determinant MyoD and functional Brg1, the ATPase subunit of SWI/SNF chromatin remodeling enzymes. Ectopic expression of myogenin and a specific Mef2 isoform induced myogenic differentiation without activating endogenous MyoD expression. Under these conditions, the regulatory sequences of late gene loci were not in close proximity, and these genes were prematurely activated. The data indicate that the spatial organization of late genes contributes to temporal regulation of myogenic transcription by restricting late gene expression during the early stages of myogenesis.
Collapse
Affiliation(s)
- Akihito Harada
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | - Chandrashekara Mallappa
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Seiji Okada
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | - John T Butler
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Stephen P Baker
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jeanne B Lawrence
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Yasuyuki Ohkawa
- Department of Advanced Medical Initiatives, JST-CREST, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Anthony N Imbalzano
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| |
Collapse
|
27
|
Kelc R, Trapecar M, Gradisnik L, Rupnik MS, Vogrin M. Platelet-rich plasma, especially when combined with a TGF-β inhibitor promotes proliferation, viability and myogenic differentiation of myoblasts in vitro. PLoS One 2015; 10:e0117302. [PMID: 25679956 PMCID: PMC4332503 DOI: 10.1371/journal.pone.0117302] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 12/21/2014] [Indexed: 01/28/2023] Open
Abstract
Regeneration of skeletal muscle after injury is limited by scar formation, slow healing time and a high recurrence rate. A therapy based on platelet-rich plasma (PRP) has become a promising lead for tendon and ligament injuries in recent years, however concerns have been raised that PRP-derived TGF-β could contribute to fibrotic remodelling in skeletal muscle after injury. Due to the lack of scientific grounds for a PRP -based muscle regeneration therapy, we have designed a study using human myogenic progenitors and evaluated the potential of PRP alone and in combination with decorin (a TGF-β inhibitor), to alter myoblast proliferation, metabolic activity, cytokine profile and expression of myogenic regulatory factors (MRFs). Advanced imaging multicolor single-cell analysis enabled us to create a valuable picture on the ratio of quiescent, activated and terminally committed myoblasts in treated versus control cell populations. Finally high-resolution confocal microscopy validated the potential of PRP and decorin to stimulate the formation of polynucleated myotubules. PRP was shown to down-regulate fibrotic cytokines, increase cell viability and proliferation, enhance the expression of MRFs, and contribute to a significant myogenic shift during differentiation. When combined with decorin further synergistc effects were identified. These results suggest that PRP could not only prevent fibrosis but could also stimulate muscle commitment, especially when combined with a TGF-β inhibitor.
Collapse
Affiliation(s)
- Robi Kelc
- Department of Orthopaedic Surgery, University Medical Center Maribor, Maribor, Slovenia
- * E-mail:
| | - Martin Trapecar
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Lidija Gradisnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Marjan Slak Rupnik
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Matjaz Vogrin
- Department of Orthopaedic Surgery, University Medical Center Maribor, Maribor, Slovenia
| |
Collapse
|
28
|
Capetanaki Y, Papathanasiou S, Diokmetzidou A, Vatsellas G, Tsikitis M. Desmin related disease: a matter of cell survival failure. Curr Opin Cell Biol 2015; 32:113-20. [PMID: 25680090 DOI: 10.1016/j.ceb.2015.01.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 01/12/2015] [Accepted: 01/21/2015] [Indexed: 12/19/2022]
Abstract
Maintenance of the highly organized striated muscle tissue requires a cell-wide dynamic network that through interactions with all vital cell structures, provides an effective mechanochemical integrator of morphology and function, absolutely necessary for intra-cellular and intercellular coordination of all muscle functions. A good candidate for such a system is the desmin intermediate filament cytoskeletal network. Human desmin mutations and post-translational modifications cause disturbance of this network, thus leading to loss of function of both desmin and its binding partners, as well as potential toxic effects of the formed aggregates. Both loss of normal function and gain of toxic function are linked to mitochondrial defects, cardiomyocyte death, muscle degeneration and development of skeletal myopathy and cardiomyopathy.
Collapse
Affiliation(s)
- Yassemi Capetanaki
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece.
| | - Stamatis Papathanasiou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Antigoni Diokmetzidou
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Giannis Vatsellas
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - Mary Tsikitis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| |
Collapse
|
29
|
Lim CJ, Jeon JE, Jeong SK, Yoon SJ, Kwon SD, Lim J, Park K, Kim DY, Ahn JK, Kim BW. Growth hormone-releasing peptide-biotin conjugate stimulates myocytes differentiation through insulin-like growth factor-1 and collagen type I. BMB Rep 2015; 48:501-6. [PMID: 25644636 PMCID: PMC4641233 DOI: 10.5483/bmbrep.2015.48.9.258] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Indexed: 11/20/2022] Open
Abstract
Based on the potential beneficial effects of growth hormone releasing peptide (GHRP)-6 on muscle functions, a newly synthesized GHRP-6-biotin conjugate was tested on cultured myoblast cells. Increased expression of myogenic marker proteins was observed in GHRP-6-biotin conjugate-treated cells. Additionally, increased expression levels of insulin-like growth factor-1 and collagen type I were observed. Furthermore, GHRP-6-biotin conjugate-treated cells showed increased metabolic activity, as indicated by increased concentrations of energy metabolites, such as ATP and lactate, and increased enzymatic activity of lactate dehydrogenase and creatine kinase. Finally, binding protein analysis suggested few candidate proteins, including desmin, actin, and zinc finger protein 691 as potential targets for GHRP6-biotin conjugate action. These results suggest that the newly synthesized GHRP-6-biotin conjugate has myogenic stimulating activity through, at least in part, by stimulating collagen type I synthesis and several key proteins. Practical applications of the GHRP-6-biotin conjugate could include improving muscle condition. [BMB Reports 2015; 48(9): 501-506]
Collapse
Affiliation(s)
- Chae Jin Lim
- Department of Microbiology and Molecular Biology, School of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea; Peptide R&D Center, Incospharm Corporation, Daejeon 34141, Korea
| | - Jung Eun Jeon
- CRID Center, NeoPharm Co., Ltd., Daejeon 34037, Korea
| | - Se Kyoo Jeong
- CRID Center, NeoPharm Co., Ltd., Daejeon 34037, Korea
| | - Seok Jeong Yoon
- Peptide R&D Center, Incospharm Corporation, Daejeon 28674, Korea
| | - Seon Deok Kwon
- Peptide R&D Center, Incospharm Corporation, Daejeon 28674, Korea
| | - Jina Lim
- Peptide R&D Center, Incospharm Corporation, Daejeon 28674, Korea
| | - Keedon Park
- Peptide R&D Center, Incospharm Corporation, Daejeon 28674, Korea
| | - Dae Yong Kim
- Departments of Pharmaceutical Science and Engineering, Seowon University, Cheongju 28674, Korea
| | - Jeong Keun Ahn
- Department of Microbiology and Molecular Biology, School of Bioscience and Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Bong-Woo Kim
- Departments of Cosmetic Science & Technology, Seowon University, Cheongju 28674, Korea
| |
Collapse
|
30
|
Hnia K, Ramspacher C, Vermot J, Laporte J. Desmin in muscle and associated diseases: beyond the structural function. Cell Tissue Res 2014; 360:591-608. [PMID: 25358400 DOI: 10.1007/s00441-014-2016-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 09/22/2014] [Indexed: 11/25/2022]
Abstract
Desmin is a muscle-specific type III intermediate filament essential for proper muscular structure and function. In human, mutations affecting desmin expression or promoting its aggregation lead to skeletal (desmin-related myopathies), or cardiac (desmin-related cardiomyopathy) phenotypes, or both. Patient muscles display intracellular accumulations of misfolded proteins and desmin-positive insoluble granulofilamentous aggregates, leading to a large spectrum of molecular alterations. Increasing evidence shows that desmin function is not limited to the structural and mechanical integrity of cells. This novel perception is strongly supported by the finding that diseases featuring desmin aggregates cannot be easily associated with mechanical defects, but rather involve desmin filaments in a broader spectrum of functions, such as in organelle positioning and integrity and in signaling. Here, we review desmin functions and related diseases affecting striated muscles. We detail emergent cellular functions of desmin based on reported phenotypes in patients and animal models. We discuss known desmin protein partners and propose an overview of the way that this molecular network could serve as a signal transduction platform necessary for proper muscle function.
Collapse
Affiliation(s)
- Karim Hnia
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France,
| | | | | | | |
Collapse
|
31
|
Treskes P, Neef K, Perumal Srinivasan S, Halbach M, Stamm C, Cowan D, Scherner M, Madershahian N, Wittwer T, Hescheler J, Wahlers T, Choi YH. Preconditioning of skeletal myoblast-based engineered tissue constructs enables functional coupling to myocardium in vivo. J Thorac Cardiovasc Surg 2014; 149:348-56. [PMID: 25439779 DOI: 10.1016/j.jtcvs.2014.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Skeletal myoblasts fuse to form functional syncytial myotubes as an integral part of the skeletal muscle. During this differentiation process, expression of proteins for mechanical and electrical integration is seized, which is a major drawback for the application of skeletal myoblasts in cardiac regenerative cell therapy, because global heart function depends on intercellular communication. METHODS Mechanically preconditioned engineered tissue constructs containing neonatal mouse skeletal myoblasts were transplanted epicardially. A Y-chromosomal specific polymerase chain reaction (PCR) was undertaken up to 10 weeks after transplantation to confirm the presence of grafted cells. Histologic and electrophysiologic analyses were carried out 1 week after transplantation. RESULTS Cells within the grafted construct expressed connexin 43 at the interface to the host myocardium, indicating electrical coupling, confirmed by sharp electrode recordings. Analyses of the maximum stimulation frequency (5.65 ± 0.37 Hz), conduction velocity (0.087 ± 0.011 m/s) and sensitivity for pharmacologic conduction block (0.736 ± 0.080 mM 1-heptanol) revealed effective electrophysiologic coupling between graft and host cells, although significantly less robust than in native myocardial tissue (maximum stimulation frequency, 11.616 ± 0.238 Hz, P < .001; conduction velocity, 0.300 ± 0.057 m/s, P < .01; conduction block, 1.983 ± 0.077 mM 1-heptanol, P < .001). CONCLUSIONS Although untreated skeletal myoblasts cannot couple to cardiomyocytes, we confirm that mechanical preconditioning enables transplanted skeletal myoblasts to functionally interact with cardiomyocytes in vivo and, thus, reinvigorate the concept of skeletal myoblast-based cardiac cell therapy.
Collapse
Affiliation(s)
- Philipp Treskes
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sureshkumar Perumal Srinivasan
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Marcel Halbach
- Institute for Neurophysiology, University of Cologne, Cologne, Germany; Department of Internal Medicine III, Heart Center, University of Cologne, Cologne, Germany
| | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Douglas Cowan
- Departments of Anesthesiology and Perioperative and Pain Medicine, Children's Hospital Boston and Harvard Medical School, Boston, Mass
| | - Maximilian Scherner
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Navid Madershahian
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Thorsten Wittwer
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
32
|
Sondag GR, Salihoglu S, Lababidi SL, Crowder DC, Moussa FM, Abdelmagid SM, Safadi FF. Osteoactivin induces transdifferentiation of C2C12 myoblasts into osteoblasts. J Cell Physiol 2014; 229:955-66. [PMID: 24265122 DOI: 10.1002/jcp.24512] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 11/18/2013] [Indexed: 12/19/2022]
Abstract
Osteoactivin (OA) is a novel osteogenic factor important for osteoblast differentiation and function. Previous studies showed that OA stimulates matrix mineralization and transcription of osteoblast specific genes required for differentiation. OA plays a role in wound healing and its expression was shown to increase in post fracture calluses. OA expression was reported in muscle as OA is upregulated in cases of denervation and unloading stress. The regulatory mechanisms of OA in muscle and bone have not yet been determined. In this study, we examined whether OA plays a role in transdifferentiation of C2C12 myoblast into osteoblasts. Infected C2C12 with a retroviral vector overexpressing OA under the CMV promoter were able to transdifferentiate from myoblasts into osteoblasts. Immunofluorescence analysis showed that skeletal muscle marker MF-20 was severely downregulated in cells overexpressing OA and contained significantly less myotubes compared to uninfected control. C2C12 myoblasts overexpressing OA showed an increase in expression of bone specific markers such as alkaline phosphatase and alizarin red staining, and also showed an increase in Runx2 protein expression. We also detected increased levels of phosphorylated focal adhesion kinase (FAK) in C2C12 myoblasts overexpressing OA compared to control. Taken together, our results suggest that OA is able to induce transdifferentiation of myoblasts into osteoblasts through increasing levels of phosphorylated FAK.
Collapse
Affiliation(s)
- Gregory R Sondag
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio; School of Biomedical Sciences, Kent State University, Kent, Ohio
| | | | | | | | | | | | | |
Collapse
|
33
|
Winter DL, Paulin D, Mericskay M, Li Z. Posttranslational modifications of desmin and their implication in biological processes and pathologies. Histochem Cell Biol 2013; 141:1-16. [DOI: 10.1007/s00418-013-1148-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2013] [Indexed: 11/29/2022]
|
34
|
Knockdown of endogenous myostatin promotes sheep myoblast proliferation. In Vitro Cell Dev Biol Anim 2013; 50:94-102. [PMID: 24052475 DOI: 10.1007/s11626-013-9689-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 09/06/2013] [Indexed: 02/06/2023]
Abstract
Myostatin (MSTN), is a known negative regulator of myogenesis. Silencing of the function of MSTN could result in increasing muscle mass in mice. To determine the function of endogenous MSTN expression on proliferation of sheep myoblasts, a short-hairpin RNA-targeting sheep MSTN was constructed into lentiviral vector to silence endogenous MSTN expression. We demonstrated that silencing of endogenous MSTN gene with up to approximately 73.3% reduction by short hairpin RNA (shRNA) resulted in significant increase (overall 28.3%) of proliferation of primary ovine myoblasts. The upregulation of proliferation was accompanied by the decrease expression of MyoD (-37.6%, p = 0.025), myogenin (-33.1%, p = 0.049), p21 (-49.3%, p = 0.046), and Smad3 (-50.0%, p = 0.007). Silencing of myostatin using shRNA may provide a feasible approach to improve meat productivity in farm animals.
Collapse
|
35
|
Tsoupri E, Capetanaki Y. Μyospryn: a multifunctional desmin-associated protein. Histochem Cell Biol 2013; 140:55-63. [PMID: 23748244 DOI: 10.1007/s00418-013-1103-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2013] [Indexed: 12/14/2022]
Abstract
Desmin, the muscle-specific intermediate filament protein, forms a 3D scaffold that links the contractile apparatus to the costameres of plasma membrane, intercalated disks, the nucleus, and also other membranous organelles. The cellular scaffold formed by desmin and its binding partners might be implicated in signaling and trafficking processes, vital mechanisms for the survival of the mammalian cell. One novel desmin-associated protein is the tripartite motif-like protein myospryn. Myospryn was initially identified as an associated partner to the biogenesis of lysosome-related organelles complex 1 protein dysbindin, implicating its potential involvement in vesicle trafficking and organelle biogenesis and/or positioning. Myospryn is also an A kinase anchoring protein, raising the possibility that together with desmin and other cytoskeletal and signaling proteins, it could participate in the subcellular targeting of protein kinase A activity in striated muscle. As with desmin, different members of this scaffold might play a crucial role in the pathogenesis of muscle disease, since any disturbance in these highly coordinated signaling pathways is expected to compromise efficient maintenance of structure-function integrity of muscle and lead to different cardiac and skeletal myopathies.
Collapse
Affiliation(s)
- Elsa Tsoupri
- Cell Biology Division, Center of Basic Research, Biomedical Research Foundation Academy of Athens, 11527, Athens, Greece
| | | |
Collapse
|
36
|
Krause MP, Moradi J, Coleman SK, D'Souza DM, Liu C, Kronenberg MS, Rowe DW, Hawke TJ, Hadjiargyrou M. A novel GFP reporter mouse reveals Mustn1 expression in adult regenerating skeletal muscle, activated satellite cells and differentiating myoblasts. Acta Physiol (Oxf) 2013; 208:180-90. [PMID: 23506283 DOI: 10.1111/apha.12099] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 03/08/2013] [Accepted: 03/12/2013] [Indexed: 11/30/2022]
Abstract
AIM Mustn1 has been implicated in myofusion as well as skeletal muscle growth and repair; however, the exact role and spatio-temporal expression of Mustn1 have yet to be fully defined. METHODS Transgenic mice were generated with a 1512-bp sequence of the Mustn1 promoter directing the expression of GFP (Mustn1(PRO) -GFP). These mice were used to investigate the spatio-temporal expression of Mustn1(PRO) -GFP during skeletal muscle development and adult skeletal muscle repair, as well as various phases of the satellite cell lifespan (i.e. quiescence, activation, proliferation, differentiation). RESULTS Mustn1(PRO) -GFP expression was observed within somites at embryonic day 12 and developing skeletal muscles at embryonic day 15 and 18. While uninjured adult tibialis anterior muscle displayed no detectable Mustn1(PRO) -GFP expression, cardiotoxin injury robustly elevated Mustn1(PRO) -GFP expression at 3 days post-injury with decreasing levels observed at 5 days and minimal, focal expression seen at 10 days. The expression of Mustn1(PRO) -GFP at 3 days post-injury consistently overlaid with MyoD although the strongest expression of Mustn1(PRO) -GFP was noted in newly formed myotubes that were expressing minimal levels of MyoD. By 5 days post-injury, Mustn1(PRO) -GFP overlaid in all myotubes expressing myogenin although cells were present expressing Mustn1(PRO) -GFP alone. The expression patterns of Mustn1(PRO) -GFP in regenerating muscle preceded the expression of desmin throughout the regenerative time course consistent with Mustn1 being upstream of this myogenic protein. Further, quiescent satellite cells located on freshly isolated, single myofibers rarely expressed Mustn1(PRO) -GFP, but within 24 h of isolation, all activated satellite cells expressed Mustn1(PRO) -GFP. Expression of Mustn1(PRO) -GFP in primary myoblasts diminished with prolonged time in proliferation media. However, in response to serum withdrawal, the expression of Mustn1(PRO) -GFP increased during myofusion (day 2) followed by declining expression thereafter. CONCLUSION Mustn1(PRO) -GFP is expressed in activated satellite cells and myoblasts but continued time in proliferation media diminished Mustn1(PRO) -GFP expression. However, myoblasts exposed to serum withdrawal increased Mustn1(PRO) -GFP expression consistent with its demonstrated role in myofusion. The in vivo expression pattern of Mustn1 observed in regenerating and developing skeletal muscle is consistent with its presence in satellite cells and its critical role in myofusion.
Collapse
Affiliation(s)
- M. P. Krause
- Department of Pathology and Molecular Medicine; McMaster University; Hamilton; Ontario; Canada
| | - J. Moradi
- Department of Pathology and Molecular Medicine; McMaster University; Hamilton; Ontario; Canada
| | - S. K. Coleman
- Department of Pathology and Molecular Medicine; McMaster University; Hamilton; Ontario; Canada
| | - D. M. D'Souza
- Department of Pathology and Molecular Medicine; McMaster University; Hamilton; Ontario; Canada
| | - C. Liu
- Department of Life Sciences; Theobald Science Center; New York Institute of Technology; Old Westbury; NY; USA
| | - M. S. Kronenberg
- Department of Genetics and Developmental Biology; University of Connecticut Health Center; Farmington; CT; USA
| | - D. W. Rowe
- Department of Genetics and Developmental Biology; University of Connecticut Health Center; Farmington; CT; USA
| | - T. J. Hawke
- Department of Pathology and Molecular Medicine; McMaster University; Hamilton; Ontario; Canada
| | - M. Hadjiargyrou
- Department of Life Sciences; Theobald Science Center; New York Institute of Technology; Old Westbury; NY; USA
| |
Collapse
|
37
|
Smoczer C, Hooker L, Brode S, Wolanski M, KhosrowShahian F, Crawford M. The Xenopus homeobox gene pitx3 impinges upon somitogenesis and laterality. Biochem Cell Biol 2013; 91:79-87. [PMID: 23527636 DOI: 10.1139/bcb-2012-0057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pitx3 has been identified as the causative locus in a developmental eye mutation associated with mammalian anterior segment dysgenesis, congenital cataracts, and aphakia. In recent studies of frog eye development we discovered that pitx3 expresses symmetrically in the somites and lateral plate mesoderm and asymmetrically during cardiac and gut looping. We report that disruption of pitx3 activity on one side of an embryo relative to the other, either by over- or underexpression of pitx3, elicits a crooked dorsal axis in embryos that is a consequence of a retarded progression through somitogenesis. Unlike in amniotes, Xenopus somites form as cohorts of presomitic cells that rotate perpendicular to the dorsal axis. Since no vertebral anomalies have been reported in mouse and human Pitx3 mutants, we attempt to distinguish whether the segmentation clock is uniquely affected in frog or if the pitx3 perturbation inhibits the cellular changes that are necessary to rotation of presomitic cells. In Xenopus, pitx3 appears to inhibit the rotation of presomitic cell cohorts and to be necessary to the bilaterally symmetric expression of pitx2 in somites.
Collapse
Affiliation(s)
- Cristine Smoczer
- Biological Science, University of Windsor, 401 Sunset Avenue, Windsor, ON N9B 3P4, Canada
| | | | | | | | | | | |
Collapse
|
38
|
Improved tissue culture conditions for engineered skeletal muscle sheets. ScientificWorldJournal 2013; 2013:370151. [PMID: 23533347 PMCID: PMC3603544 DOI: 10.1155/2013/370151] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Accepted: 01/24/2013] [Indexed: 12/22/2022] Open
Abstract
The potential clinical utility of engineered muscle is currently restricted by limited in vitro capacity of expanded muscle precursor cells to fuse and form mature myofibers. The purpose of this study was to use isotropic skeletal muscle sheets to explore the impact of (1) fibroblast coculture and (2) fibroblast-conditioned media (fCM) on in vitro myogenesis. Muscle sheets were prepared by seeding varying ratios of skeletal myoblasts and fibroblasts on a biomimetic substrate and culturing the resulting tissue in either control media or fCM. Muscle sheets were prepared from two cell subpopulations, (1) C2C12 and NOR-10 and (2) primary neonatal rat skeletal muscle cells (nSKM). In C2C12/Nor-10 muscle sheets fCM conferred a myogenic advantage early in culture; at D1 a statistically significant 3.12 ± 0.8-fold increase in myofiber density was observed with fCM. A high purity satellite cell population was collected from an initially mixed population of nSKMs via cell sorting for positive α 7-integrin expression. On D6, tissue sheets with low fibroblast concentrations (0 & 10%) cultured in fCM had increased average myofiber density (4.8 ± 0.2 myofibers/field) compared to tissue sheets with high fibroblast concentrations (50%) cultured in control media (1.0 ± 0.1 myofibers/field). Additionally, fCM promoted longer, thicker myofibers with a mature phenotype.
Collapse
|
39
|
Desminopathies: pathology and mechanisms. Acta Neuropathol 2013; 125:47-75. [PMID: 23143191 PMCID: PMC3535371 DOI: 10.1007/s00401-012-1057-6] [Citation(s) in RCA: 162] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2012] [Revised: 10/15/2012] [Accepted: 10/18/2012] [Indexed: 12/22/2022]
Abstract
The intermediate filament protein desmin is an essential component of the extra-sarcomeric cytoskeleton in muscle cells. This three-dimensional filamentous framework exerts central roles in the structural and functional alignment and anchorage of myofibrils, the positioning of cell organelles and signaling events. Mutations of the human desmin gene on chromosome 2q35 cause autosomal dominant, autosomal recessive, and sporadic myopathies and/or cardiomyopathies with marked phenotypic variability. The disease onset ranges from childhood to late adulthood. The clinical course is progressive and no specific treatment is currently available for this severely disabling disease. The muscle pathology is characterized by desmin-positive protein aggregates and degenerative changes of the myofibrillar apparatus. The molecular pathophysiology of desminopathies is a complex, multilevel issue. In addition to direct effects on the formation and maintenance of the extra-sarcomeric intermediate filament network, mutant desmin affects essential protein interactions, cell signaling cascades, mitochondrial functions, and protein quality control mechanisms. This review summarizes the currently available data on the epidemiology, clinical phenotypes, myopathology, and genetics of desminopathies. In addition, this work provides an overview on the expression, filament formation processes, biomechanical properties, post-translational modifications, interaction partners, subcellular localization, and functions of wild-type and mutant desmin as well as desmin-related cell and animal models.
Collapse
|
40
|
Elowsson L, Kirsebom H, Carmignac V, Mattiasson B, Durbeej M. Evaluation of macroporous blood and plasma scaffolds for skeletal muscle tissue engineering. Biomater Sci 2013; 1:402-410. [DOI: 10.1039/c2bm00054g] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
41
|
Kammoun M, Picard B, Henry-Berger J, Cassar-Malek I. A network-based approach for predicting Hsp27 knock-out targets in mouse skeletal muscles. Comput Struct Biotechnol J 2013; 6:e201303008. [PMID: 24688716 PMCID: PMC3962151 DOI: 10.5936/csbj.201303008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/04/2013] [Accepted: 07/10/2013] [Indexed: 12/16/2022] Open
Abstract
Thanks to genomics, we have previously identified markers of beef tenderness, and computed a bioinformatic analysis that enabled us to build an interactome in which we found Hsp27 at a crucial node. Here, we have used a network-based approach for understanding the contribution of Hsp27 to tenderness through the prediction of its interactors related to tenderness. We have revealed the direct interactors of Hsp27. The predicted partners of Hsp27 included proteins involved in different functions, e.g. members of Hsp families (Hsp20, Cryab, Hsp70a1a, and Hsp90aa1), regulators of apoptosis (Fas, Chuk, and caspase-3), translation factors (Eif4E, and Eif4G1), cytoskeletal proteins (Desmin) and antioxidants (Sod1). The abundances of 15 proteins were quantified by Western blotting in two muscles of HspB1-null mice and their controls. We observed changes in the amount of most of the Hsp27 predicted targets in mice devoid of Hsp27 mainly in the most oxidative muscle. Our study demonstrates the functional links between Hsp27 and its predicted targets. It suggests that Hsp status, apoptotic processes and protection against oxidative stress are crucial for post-mortem muscle metabolism, subsequent proteolysis, and therefore for beef tenderness.
Collapse
Affiliation(s)
- Malek Kammoun
- INRA, UMR1213 Herbivores, F-63122 Saint-Genès-Champanelle, France
- Clermont University, VetAgro Sup, UMR1213 Herbivores, BP 10448, F-63000, Clermont-Ferrand, France
| | - Brigitte Picard
- INRA, UMR1213 Herbivores, F-63122 Saint-Genès-Champanelle, France
- Clermont University, VetAgro Sup, UMR1213 Herbivores, BP 10448, F-63000, Clermont-Ferrand, France
| | | | - Isabelle Cassar-Malek
- INRA, UMR1213 Herbivores, F-63122 Saint-Genès-Champanelle, France
- Clermont University, VetAgro Sup, UMR1213 Herbivores, BP 10448, F-63000, Clermont-Ferrand, France
- Corresponding author: E-mail address: (Isabelle Cassar-Malek)
| |
Collapse
|
42
|
The critical role of myostatin in differentiation of sheep myoblasts. Biochem Biophys Res Commun 2012; 422:381-6. [DOI: 10.1016/j.bbrc.2012.04.151] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 04/27/2012] [Indexed: 12/27/2022]
|
43
|
Berger F, Büchsler I, Munz B. The effect of the NF-kappa B inhibitors curcumin and lactacystin on myogenic differentiation of rhabdomyosarcoma cells. Differentiation 2012; 83:271-81. [PMID: 22469857 DOI: 10.1016/j.diff.2012.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 01/23/2012] [Accepted: 02/01/2012] [Indexed: 11/17/2022]
Abstract
Rhabdomyosarcoma is a soft tissue sarcoma mainly seen in children. Despite considerable progress within the last few years, therapeutic approaches for this type of tumor are still limited. The respective tumor cells originate from myogenic precursor cells and are characterized by a blockade in their differentiation program. Interestingly, there is a direct inverse correlation between the differentiation status of a specific rhabdomyosarcoma cell and its metastatic potential. Thus, here, we tested whether the ubiquitous transcription factor NF-κB, which regulates myogenic differentiation and is also a promising therapeutic target in the treatment of other types of tumors, might be an interesting candidate for the development of novel rhabdomyosarcoma treatment strategies. For this purpose, we analyzed NF-κB activity (classical pathway) in myoblasts with different differentiation potential, specifically in three different rhabdomyosarcoma cell lines. In addition, we inhibited NF-κB activity in these cells and analyzed the effects on myogenic differentiation. We show that after the induction of differentiation, NF-κB activity declines rapidly in normal myoblasts, but only slightly in rhabdomyosarcoma cells. However, after treatment of the cells with two different small-molecule NF-κB-inhibiting compounds, the IKK inhibitor curcumin and the proteasome inhibitor lactacystin, we found that neither curcumin nor lactacystin promoted myogenic differentiation in either normal myoblasts or rhabdomyosarcoma cells. Taken together, our data suggest that treatment with curcumin or lactacystin might not be a suitable approach in the treatment of rhabdomyosarcoma.
Collapse
Affiliation(s)
- Felicitas Berger
- Charité, Institute of Physiology, University Medicine Berlin, Thielallee 71, D-14195 Berlin, Germany
| | | | | |
Collapse
|
44
|
Han D, Zhao H, Parada C, Hacia JG, Bringas P, Chai Y. A TGFβ-Smad4-Fgf6 signaling cascade controls myogenic differentiation and myoblast fusion during tongue development. Development 2012; 139:1640-50. [PMID: 22438570 DOI: 10.1242/dev.076653] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The tongue is a muscular organ and plays a crucial role in speech, deglutition and taste. Despite the important physiological functions of the tongue, little is known about the regulatory mechanisms of tongue muscle development. TGFβ family members play important roles in regulating myogenesis, but the functional significance of Smad-dependent TGFβ signaling in regulating tongue skeletal muscle development remains unclear. In this study, we have investigated Smad4-mediated TGFβ signaling in the development of occipital somite-derived myogenic progenitors during tongue morphogenesis through tissue-specific inactivation of Smad4 (using Myf5-Cre;Smad4(flox/flox) mice). During the initiation of tongue development, cranial neural crest (CNC) cells occupy the tongue buds before myogenic progenitors migrate into the tongue primordium, suggesting that CNC cells play an instructive role in guiding tongue muscle development. Moreover, ablation of Smad4 results in defects in myogenic terminal differentiation and myoblast fusion. Despite compromised muscle differentiation, tendon formation appears unaffected in the tongue of Myf5-Cre;Smad4(flox/flox) mice, suggesting that the differentiation and maintenance of CNC-derived tendon cells are independent of Smad4-mediated signaling in myogenic cells in the tongue. Furthermore, loss of Smad4 results in a significant reduction in expression of several members of the FGF family, including Fgf6 and Fgfr4. Exogenous Fgf6 partially rescues the tongue myoblast fusion defect of Myf5-Cre;Smad4(flox/flox) mice. Taken together, our study demonstrates that a TGFβ-Smad4-Fgf6 signaling cascade plays a crucial role in myogenic cell fate determination and lineage progression during tongue myogenesis.
Collapse
Affiliation(s)
- Dong Han
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | |
Collapse
|
45
|
Herpesviruses and intermediate filaments: close encounters with the third type. Viruses 2011; 3:1015-40. [PMID: 21994768 PMCID: PMC3185793 DOI: 10.3390/v3071015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 06/07/2011] [Accepted: 06/24/2011] [Indexed: 01/29/2023] Open
Abstract
Intermediate filaments (IF) are essential to maintain cellular and nuclear integrity and shape, to manage organelle distribution and motility, to control the trafficking and pH of intracellular vesicles, to prevent stress-induced cell death, and to support the correct distribution of specific proteins. Because of this, IF are likely to be targeted by a variety of pathogens, and may act in favor or against infection progress. As many IF functions remain to be identified, however, little is currently known about these interactions. Herpesviruses can infect a wide variety of cell types, and are thus bound to encounter the different types of IF expressed in each tissue. The analysis of these interrelationships can yield precious insights into how IF proteins work, and into how viruses have evolved to exploit these functions. These interactions, either known or potential, will be the focus of this review.
Collapse
|
46
|
Pinto A, Speckmann B, Heisler M, Sies H, Steinbrenner H. Delaying of insulin signal transduction in skeletal muscle cells by selenium compounds. J Inorg Biochem 2011; 105:812-20. [PMID: 21497580 DOI: 10.1016/j.jinorgbio.2011.03.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2011] [Revised: 03/15/2011] [Accepted: 03/15/2011] [Indexed: 11/30/2022]
Abstract
Supranutritional selenium (Se) intake and high serum Se levels have been associated epidemiologically with increased risk for type 2 diabetes, suggesting adverse effects of dietary Se compounds and/or antioxidant selenoenzymes on the sensitivity of target tissues for insulin. Here, we compared the capability of inorganic (sodium selenite and sodium selenate) and organic (selenomethionine and methylseleninic acid (MSeA)) Se compounds to interfere with insulin signaling in rat L6 myotubes, differentiated skeletal muscle cells. When applied at doses of 1 μM, only selenite and MSeA were capable of delaying insulin-induced phosphorylation of protein kinase B (Akt) and attenuating insulin-induced phosphorylation of forkhead box class O transcription factors FoxO1a and FoxO3. Insulin-stimulated glucose uptake was lowered by selenite and MSeA as well. Even though all tested Se compounds strongly stimulated expression/activity of the cellular selenoproteins glutathione peroxidase 1 and selenoprotein W, selenite and MSeA were the most efficiently utilized Se donors. Moreover, at doses of 1 μM, only selenite and MSeA had a significant inhibitory effect on generation of intracellular reactive oxygen species (ROS). These results suggest that the Se(IV) compounds selenite and MSeA may impair the insulin sensitivity of myocytes by influencing cellular redox homeostasis.
Collapse
Affiliation(s)
- Antonio Pinto
- Institute for Biochemistry and Molecular Biology I, Heinrich-Heine-University, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
47
|
Lee DH, Ro H. Effect of Exercise on Intermediate Filament Expression in the Skeletal Muscles of Rats with Sciatic Nerve Injury. J Phys Ther Sci 2011. [DOI: 10.1589/jpts.23.213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Dae-Hee Lee
- Department of Physical Therapy, Youngdong University
| | - Hyolyun Ro
- Department of Occupational Therapy, Kangwon National University
| |
Collapse
|
48
|
Lee DM, Lee KH, Cheon YP, Chun TH, Choi IH. Effects of Anabolic Steroids of Pork on Proliferation and Differentiation of Myogenic Satellite Cell. Korean J Food Sci Anim Resour 2010. [DOI: 10.5851/kosfa.2010.30.5.842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
49
|
Woo JH, Kim MJ, Kim HS. Phosphoinositide 3-kinase regulates myogenin expression at both the transcriptional and post-transcriptional level during myogenesis. Anim Cells Syst (Seoul) 2010. [DOI: 10.1080/19768354.2010.496541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
50
|
Liu C, Gersch RP, Hawke TJ, Hadjiargyrou M. Silencing of Mustn1 inhibits myogenic fusion and differentiation. Am J Physiol Cell Physiol 2010; 298:C1100-8. [PMID: 20130207 DOI: 10.1152/ajpcell.00553.2009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mustn1 (Mustang, musculoskeletal temporally activated novel gene) was originally identified in fracture callus tissue, but its greatest expression is detected in skeletal muscle. Thus, we conducted experiments to investigate the expression and function of Mustn1 during myogenesis. Temporally, quantitative real-time PCR analysis of muscle samples from embryonic day 17 to 12 mo of age reveals that Mustn1 mRNA expression is greatest at 3 mo of age and beyond, consistent with the expression pattern of Myod. In situ hybridization shows abundant Mustn1 expression in somites and developing skeletal muscles, while in adult muscle, Mustn1 is localized to some peripherally located nuclei. Using RNA interference (RNAi), we investigated the function of Mustn1 in C2C12 myoblasts. Though silencing Mustn1 mRNA had no effect on myoblast proliferation, it did significantly impair myoblast differentiation, preventing myofusion. Specifically, when placed in low-serum medium for up to 6 days, Mustn1-silenced myoblasts elongated poorly and were mononucleated. In contrast, control RNAi-treated and parental myoblasts presented as large, multinucleated myotubes. Further supporting the morphological observations, immunocytochemistry of Mustn1-silenced cells demonstrated significant reductions in myogenin (Myog) and myosin heavy chain (Myhc) expression at 4 and 6 days of differentiation as compared with control and parental cells. The decreases in Myog and Myhc protein expression in Mustn1-silenced cells were associated with robust ( approximately 3-fold or greater) decreases in the expression of Myod and desmin (Des), as well as the myofusion markers calpain 1 (Capn1), caveolin 3 (Cav3), and cadherin 15 (M-cadherin; Cadh15). Overall, we demonstrate that Mustn1 is an essential regulator of myogenic differentiation and myofusion, and our findings implicate Myod and Myog as its downstream targets.
Collapse
Affiliation(s)
- Cheng Liu
- Dept. of Biomedical Engineering, Stony Brook Univ., NY 11794-2580, USA
| | | | | | | |
Collapse
|