1
|
Rexius-Hall ML, Madrigal MD, Kilic CY, Shen K, McCain ML. Profiling paracrine interactions between hypoxic and normoxic skeletal muscle tissue in a microphysiological system fabricated from 3D printed components. LAB ON A CHIP 2025; 25:212-224. [PMID: 39665980 PMCID: PMC11887996 DOI: 10.1039/d4lc00603h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Disrupted blood flow in conditions such as peripheral artery disease and critical limb ischemia leads to variations in oxygen supply within skeletal muscle tissue, creating regions of poorly perfused, hypoxic skeletal muscle surrounded by regions of adequately perfused, normoxic muscle tissue. These oxygen gradients may have significant implications for muscle injury or disease, as mediated by the exchange of paracrine factors between differentially oxygenated tissue. However, creating and maintaining heterogeneous oxygen landscapes within a controlled experimental setup to ensure continuous paracrine signaling is a technological challenge. Here, we engineer oxygen-controlled microphysiological systems to investigate paracrine interactions between differentially oxygenated engineered muscle tissue. We fabricated microphysiological systems with dual oxygen landscapes that also had engineered control over paracrine interactions between hypoxic and normoxic skeletal muscle tissues, which were differentiated from C2C12 myoblasts cultured on micromolded gelatin hydrogels. The microphysiological systems interfaced with a new 3D-printed oxygen control well plate insert, which we designed to distribute flow to multiple microphysiological systems and minimize evaporation for longer timepoints. With our system, we demonstrated that amphiregulin, a myokine associated with skeletal muscle injury, exhibits unique upregulation in both gene expression and secretion after 24 hours due to paracrine interactions between hypoxic and normoxic skeletal muscle tissue. Our platform can be extended to investigate other impacts of paracrine interactions between hypoxic and normoxic skeletal muscle and can more broadly be used to elucidate many forms of oxygen-dependent crosstalk in other organ systems.
Collapse
Affiliation(s)
- Megan L Rexius-Hall
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA.
| | - Malinda D Madrigal
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA.
| | - Cem Y Kilic
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA.
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA.
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Megan L McCain
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, DRB 140, Los Angeles, CA 90089, USA.
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
2
|
Jung U, Kim M, Dowker-Key P, Noë S, Bettaieb A, Shepherd E, Voy B. Hypoxia promotes proliferation and inhibits myogenesis in broiler satellite cells. Poult Sci 2024; 103:103203. [PMID: 37980759 PMCID: PMC10685027 DOI: 10.1016/j.psj.2023.103203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/07/2023] [Accepted: 10/12/2023] [Indexed: 11/21/2023] Open
Abstract
Breast muscle myopathies in broilers compromise meat quality and continue to plague the poultry industry. Broiler breast muscle myopathies are characterized by impaired satellite cell (SC)-mediated repair, and localized tissue hypoxia and dysregulation of oxygen homeostasis have been implicated as contributing factors. The present study was designed to test the hypothesis that hypoxia disrupts the ability of SC to differentiate and form myotubes, both of which are key components of myofiber repair, and to determine the extent to which effects are reversed by restoration of oxygen tension. Primary SC were isolated from pectoralis major of young (5 d) Cobb 700 chicks and maintained in growth conditions or induced to differentiate under normoxic (20% O2) or hypoxic (1% O2) conditions for up to 48 h. Hypoxia enhanced SC proliferation while inhibiting myogenic potential, with decreased fusion index and suppressed myotube formation. Reoxygenation after hypoxia partially reversed effects on both proliferation and myogenesis. Western blotting showed that hypoxia diminished myogenin expression, activated AMPK, upregulated proliferation markers, and increased molecular signaling of cellular stress. Hypoxia also promoted accumulation of lipid droplets in myotubes. Targeted RNAseq identified numerous differentially expressed genes across differentiation under hypoxia, including several genes that have been associated with myopathies in vivo. Altogether, these data demonstrate localized hypoxia may influence SC behavior in ways that disrupt muscle repair and promote the formation of myopathies in broilers.
Collapse
Affiliation(s)
- Usuk Jung
- Department of Animal Science, The University of Tennessee, Knoxville, TN 37996, USA
| | - Minjeong Kim
- Department of Animal Science, The University of Tennessee, Knoxville, TN 37996, USA
| | - Presley Dowker-Key
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Simon Noë
- Research Group for Neurorehabilitation (eNRGy), Department of Rehabilitation Sciences, KU Leuven, 3001 Leuven, Belgium
| | - Ahmed Bettaieb
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Elizabeth Shepherd
- Department of Animal Science, The University of Tennessee, Knoxville, TN 37996, USA
| | - Brynn Voy
- Department of Animal Science, The University of Tennessee, Knoxville, TN 37996, USA.
| |
Collapse
|
3
|
Nguyen TH, Paprzycki L, Legrand A, Declèves AE, Heher P, Limpens M, Belayew A, Banerji CRS, Zammit PS, Tassin A. Hypoxia enhances human myoblast differentiation: involvement of HIF1α and impact of DUX4, the FSHD causal gene. Skelet Muscle 2023; 13:21. [PMID: 38104132 PMCID: PMC10724930 DOI: 10.1186/s13395-023-00330-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 10/20/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Hypoxia is known to modify skeletal muscle biological functions and muscle regeneration. However, the mechanisms underlying the effects of hypoxia on human myoblast differentiation remain unclear. The hypoxic response pathway is of particular interest in patients with hereditary muscular dystrophies since many present respiratory impairment and muscle regeneration defects. For example, an altered hypoxia response characterizes the muscles of patients with facioscapulohumeral dystrophy (FSHD). METHODS We examined the impact of hypoxia on the differentiation of human immortalized myoblasts (LHCN-M2) cultured in normoxia (PO2: 21%) or hypoxia (PO2: 1%). Cells were grown in proliferation (myoblasts) or differentiation medium for 2 (myocytes) or 4 days (myotubes). We evaluated proliferation rate by EdU incorporation, used myogenin-positive nuclei as a differentiation marker for myocytes, and determined the fusion index and myosin heavy chain-positive area in myotubes. The contribution of HIF1α was studied by gain (CoCl2) and loss (siRNAs) of function experiments. We further examined hypoxia in LHCN-M2-iDUX4 myoblasts with inducible expression of DUX4, the transcription factor underlying FSHD pathology. RESULTS We found that the hypoxic response did not impact myoblast proliferation but activated precocious myogenic differentiation and that HIF1α was critical for this process. Hypoxia also enhanced the late differentiation of human myocytes, but in an HIF1α-independent manner. Interestingly, the impact of hypoxia on muscle cell proliferation was influenced by dexamethasone. In the FSHD pathological context, DUX4 suppressed HIF1α-mediated precocious muscle differentiation. CONCLUSION Hypoxia stimulates myogenic differentiation in healthy myoblasts, with HIF1α-dependent early steps. In FSHD, DUX4-HIF1α interplay indicates a novel mechanism by which DUX4 could interfere with HIF1α function in the myogenic program and therefore with FSHD muscle performance and regeneration.
Collapse
Affiliation(s)
- Thuy-Hang Nguyen
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, Mons, 7000, Belgium
| | - Lise Paprzycki
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, Mons, 7000, Belgium
| | - Alexandre Legrand
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, Mons, 7000, Belgium
| | - Anne-Emilie Declèves
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, Mons, 7000, Belgium
| | - Philipp Heher
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Maelle Limpens
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, Mons, 7000, Belgium
| | - Alexandra Belayew
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, Mons, 7000, Belgium
| | - Christopher R S Banerji
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
- The Alan Turing Institute, British Library, 96 Euston Rd, London, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Alexandra Tassin
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, Mons, 7000, Belgium.
| |
Collapse
|
4
|
Jatwani A, Tulsawani R. Ganoderma lucidum Induces Myogenesis Markers to Avert Damage to Skeletal Muscles in Rats Exposed to Hypobaric Hypoxia. High Alt Med Biol 2023; 24:287-295. [PMID: 34142874 DOI: 10.1089/ham.2020.0172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Jatwani, Arti, and Rajkumar Tulsawani. Ganoderma lucidum induces myogenesis markers to avert damage to skeletal muscles in rats exposed to hypobaric hypoxia. High Alt Med Biol. 24:287-295, 2023. Background: Hypobaric hypoxia (HH) has been reported to induce skeletal muscle loss and impair myogenesis. Aqueous extract of G. lucidum (AqGL) contains bioactive metabolites attributed to various pharmacological effects. In this study, protective effect of AqGL in ameliorating muscle mass loss following acute HH has been reported. Materials and Methods: Male Sprague-Dawley rats were divided into following five groups of six rats in each group: unexposed control (Group 1), 6 hours of HH exposure (Group 2), 6 hours of HH exposure+AqGL extract 50 mg/kg body weight (BW) (Group 3), 6 hours of HH exposure+AqGL extract 100 mg/kg BW (Group 4), and 6 hours of HH exposure+AqGL extract 200 mg/kg BW (Group 5). Experimental animals from all groups, except Group, 1 were exposed to HH, simulated altitude of 25,000 ft for 6 hours. After exposure period, gastrocnemius muscle was collected, weighed, and morphological, biochemical, and molecular markers were analyzed. Results: HH-exposed rat muscle showed significant (p < 0.05) increase in oxidative stress markers (reactive oxygen species & malondialdehyde), which was concomitant with decrease in its mass compared to controls. AqGL treatment significantly (p < 0.05) prevented muscle oxidative stress, restored reduced glutathione content, reduced protein carbonyl content and advanced oxidation protein product, and restored muscle mass loss at effective dose of 100 mg/kg BW. Furthermore, AqGL supplementation enhanced Myf5 (p < 0.01), MyoD (p < 0.01), MyoG (p < 0.05), and Mrf4 (nonsignificantly), brain-derived neurotrophic factor (p < 0.01), and interleukin 6 (p < 0.01) expression along with restoration of tumor necrosis factor alpha (p < 0.001) and myostatin (p < 0.05) in hypoxia-exposed muscle, evidencing induction of myogenesis markers. Moreover, histological analysis showed increased myocyte number; nuclei shifted toward the periphery in the treatment group supporting muscle regeneration. Conclusion: AqGL supplementation attenuates muscle mass loss by preventing oxidative stress and inducing modulation in myogenesis markers under HH environment.
Collapse
Affiliation(s)
- Arti Jatwani
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | | |
Collapse
|
5
|
Lim S, Lee DE, Morena da Silva F, Koopmans PJ, Vechetti IJ, von Walden F, Greene NP, Murach KA. MicroRNA control of the myogenic cell transcriptome and proteome: the role of miR-16. Am J Physiol Cell Physiol 2023; 324:C1101-C1109. [PMID: 36971422 PMCID: PMC10191132 DOI: 10.1152/ajpcell.00071.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023]
Abstract
MicroRNAs (miRs) control stem cell biology and fate. Ubiquitously expressed and conserved miR-16 was the first miR implicated in tumorigenesis. miR-16 is low in muscle during developmental hypertrophy and regeneration. It is enriched in proliferating myogenic progenitor cells but is repressed during differentiation. The induction of miR-16 blocks myoblast differentiation and myotube formation, whereas knockdown enhances these processes. Despite a central role for miR-16 in myogenic cell biology, how it mediates its potent effects is incompletely defined. In this investigation, global transcriptomic and proteomic analyses after miR-16 knockdown in proliferating C2C12 myoblasts revealed how miR-16 influences myogenic cell fate. Eighteen hours after miR-16 inhibition, ribosomal protein gene expression levels were higher relative to control myoblasts and p53 pathway-related gene abundance was lower. At the protein level at this same time point, miR-16 knockdown globally upregulated tricarboxylic acid (TCA) cycle proteins while downregulating RNA metabolism-related proteins. miR-16 inhibition induced specific proteins associated with myogenic differentiation such as ACTA2, EEF1A2, and OPA1. We extend prior work in hypertrophic muscle tissue and show that miR-16 is lower in mechanically overloaded muscle in vivo. Our data collectively point to how miR-16 is implicated in aspects of myogenic cell differentiation. A deeper understanding of the role of miR-16 in myogenic cells has consequences for muscle developmental growth, exercise-induced hypertrophy, and regenerative repair after injury, all of which involve myogenic progenitors.
Collapse
Affiliation(s)
- Seongkyun Lim
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - David E Lee
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Francielly Morena da Silva
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
| | - Pieter J Koopmans
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| | - Ivan J Vechetti
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| | - Ferdinand von Walden
- Neuropediatrics, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas P Greene
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| | - Kevin A Murach
- Department of Health, Human Performance, and Recreation, Exercise Science Research Center, University of Arkansas, Fayetteville, Arkansas, United States
- Cell and Molecular Biology Graduate Program, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
6
|
Henrot P, Dupin I, Schilfarth P, Esteves P, Blervaque L, Zysman M, Gouzi F, Hayot M, Pomiès P, Berger P. Main Pathogenic Mechanisms and Recent Advances in COPD Peripheral Skeletal Muscle Wasting. Int J Mol Sci 2023; 24:ijms24076454. [PMID: 37047427 PMCID: PMC10095391 DOI: 10.3390/ijms24076454] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a worldwide prevalent respiratory disease mainly caused by tobacco smoke exposure. COPD is now considered as a systemic disease with several comorbidities. Among them, skeletal muscle dysfunction affects around 20% of COPD patients and is associated with higher morbidity and mortality. Although the histological alterations are well characterized, including myofiber atrophy, a decreased proportion of slow-twitch myofibers, and a decreased capillarization and oxidative phosphorylation capacity, the molecular basis for muscle atrophy is complex and remains partly unknown. Major difficulties lie in patient heterogeneity, accessing patients' samples, and complex multifactorial process including extrinsic mechanisms, such as tobacco smoke or disuse, and intrinsic mechanisms, such as oxidative stress, hypoxia, or systemic inflammation. Muscle wasting is also a highly dynamic process whose investigation is hampered by the differential protein regulation according to the stage of atrophy. In this review, we report and discuss recent data regarding the molecular alterations in COPD leading to impaired muscle mass, including inflammation, hypoxia and hypercapnia, mitochondrial dysfunction, diverse metabolic changes such as oxidative and nitrosative stress and genetic and epigenetic modifications, all leading to an impaired anabolic/catabolic balance in the myocyte. We recapitulate data concerning skeletal muscle dysfunction obtained in the different rodent models of COPD. Finally, we propose several pathways that should be investigated in COPD skeletal muscle dysfunction in the future.
Collapse
Affiliation(s)
- Pauline Henrot
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Isabelle Dupin
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
| | - Pierre Schilfarth
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Pauline Esteves
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
| | - Léo Blervaque
- PhyMedExp, INSERM-CNRS-Montpellier University, F-34090 Montpellier, France
| | - Maéva Zysman
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| | - Fares Gouzi
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, F-34090 Montpellier, France
| | - Maurice Hayot
- PhyMedExp, INSERM-CNRS-Montpellier University, CHRU Montpellier, F-34090 Montpellier, France
| | - Pascal Pomiès
- PhyMedExp, INSERM-CNRS-Montpellier University, F-34090 Montpellier, France
| | - Patrick Berger
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ. Bordeaux, U1045, F-33604 Pessac, France
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, CIC 1401, F-33604 Pessac, France
- CHU de Bordeaux, Service d'Exploration Fonctionnelle Respiratoire, CIC 1401, Service de Pneumologie, F-33604 Pessac, France
| |
Collapse
|
7
|
Ock SA, Seo KM, Ju WS, Kim YI, Wi HY, Lee P. Effect of Serum and Oxygen on the In Vitro Culture of Hanwoo Korean Native Cattle-Derived Skeletal Myogenic Cells Used in Cellular Agriculture. Foods 2023; 12:foods12071384. [PMID: 37048206 PMCID: PMC10093918 DOI: 10.3390/foods12071384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Skeletal muscle-derived myogenic cells (SKMCs) are novel protein sources capable of replacing animal meat. However, SKMCs have not been commercialized owing to poor productivity and the high cost of in vitro cell culture. Therefore, we cultured SKMCs in varying serum (5–20%) and oxygen concentrations (5–20%) to investigate the parameters that most impact cell productivity (serum, hypoxia, and culture medium) and examined cell proliferation ability and genes involved in myogenesis/proliferation/apoptosis/reactive oxygen species (ROS). In fetal bovine serum (FBS) groups, hypoxia induction doubled cell number, and the 20% FBS/normoxia group exhibited similar cell numbers as 5% FBS/5% hypoxia, confirming that 5% hypoxia reduced serum requirement by four-fold. The use of 20% FBS downregulated MTF5/MYOD1/MYOG/MYH1, whereas hypoxia induction with ≤10% FBS upregulated them. Although 20% FBS lowered TERT expression through rapid cell proliferation, NOX1, a major factor of ROS, was suppressed. DMEM/F12 demonstrated better differentiation potential than F10 by upregulating MYF3/MYOD1/MYOG/MYH1 and downregulating MSTN, particularly DMEM/F12 with 2% FBS/5% hypoxia. The myogenic fusion index was higher in DMEM/F12 without FBS than in DMEM/F12 with FBS (0.5–5%); however, the total nuclei number was reduced owing to apoptosis. Therefore, high serum levels are essential in influencing SKMC growth, followed by hypoxia as a synergistic component.
Collapse
|
8
|
Łoboda A, Dulak J. Nuclear Factor Erythroid 2-Related Factor 2 and Its Targets in Skeletal Muscle Repair and Regeneration. Antioxid Redox Signal 2023; 38:619-642. [PMID: 36597355 DOI: 10.1089/ars.2022.0208] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Significance: Skeletal muscles have a robust regenerative capacity in response to acute and chronic injuries. Muscle repair and redox homeostasis are intimately linked; increased generation of reactive oxygen species leads to cellular dysfunction and contributes to muscle wasting and progression of muscle diseases. In exemplary muscle disease, Duchenne muscular dystrophy (DMD), caused by mutations in the DMD gene that encodes the muscle structural protein dystrophin, the regeneration machinery is severely compromised, while oxidative stress contributes to the progression of the disease. The nuclear factor erythroid 2-related factor 2 (NRF2) and its target genes, including heme oxygenase-1 (HO-1), provide protective mechanisms against oxidative insults. Recent Advances: Relevant advances have been evolving in recent years in understanding the mechanisms by which NRF2 regulates processes that contribute to effective muscle regeneration. To this end, pathways related to muscle satellite cell differentiation, oxidative stress, mitochondrial metabolism, inflammation, fibrosis, and angiogenesis have been studied. The regulatory role of NRF2 in skeletal muscle ferroptosis has been also suggested. Animal studies have shown that NRF2 pathway activation can stop or reverse skeletal muscle pathology, especially when endogenous stress defence mechanisms are imbalanced. Critical Issues: Despite the growing recognition of NRF2 as a factor that regulates various aspects of muscle regeneration, the mechanistic impact on muscle pathology in various models of muscle injury remains imprecise. Future Directions: Further studies are necessary to fully uncover the role of NRF2 in muscle regeneration, both in physiological and pathological conditions, and to investigate the possibilities for development of new therapeutic modalities. Antioxid. Redox Signal. 38, 619-642.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Kraków, Poland
| |
Collapse
|
9
|
Silva CC, Bichara CNC, Carneiro FRO, Palacios VRDCM, den Berg AVSV, Quaresma JAS, Magno Falcão LF. Muscle dysfunction in the long coronavirus disease 2019 syndrome: Pathogenesis and clinical approach. Rev Med Virol 2022; 32:e2355. [PMID: 35416359 PMCID: PMC9111061 DOI: 10.1002/rmv.2355] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/20/2022] [Accepted: 04/01/2022] [Indexed: 01/08/2023]
Abstract
In long coronavirus disease 2019 (long COVID-19), involvement of the musculoskeletal system is characterised by the persistence or appearance of symptoms such as fatigue, muscle weakness, myalgia, and decline in physical and functional performance, even at 4 weeks after the onset of acute symptoms of COVID-19. Muscle injury biomarkers are altered during the acute phase of the disease. The cellular damage and hyperinflammatory state induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection may contribute to the persistence of symptoms, hypoxaemia, mitochondrial damage, and dysregulation of the renin-angiotensin system. In addition, the occurrence of cerebrovascular diseases, involvement of the peripheral nervous system, and harmful effects of hospitalisation, such as the use of drugs, immobility, and weakness acquired in the intensive care unit, all aggravate muscle damage. Here, we review the multifactorial mechanisms of muscle tissue injury, aggravating conditions, and associated sequelae in long COVID-19.
Collapse
Affiliation(s)
- Camilla Costa Silva
- Center for Biological and Health SciencesState University of ParaBelémBrazil
| | | | | | | | | | | | | |
Collapse
|
10
|
Bronisz-Budzyńska I, Kozakowska M, Pietraszek-Gremplewicz K, Madej M, Józkowicz A, Łoboda A, Dulak J. NRF2 Regulates Viability, Proliferation, Resistance to Oxidative Stress, and Differentiation of Murine Myoblasts and Muscle Satellite Cells. Cells 2022; 11:cells11203321. [PMID: 36291188 PMCID: PMC9600498 DOI: 10.3390/cells11203321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/27/2022] Open
Abstract
Increased oxidative stress can slow down the regeneration of skeletal muscle and affect the activity of muscle satellite cells (mSCs). Therefore, we evaluated the role of the NRF2 transcription factor (encoded by the Nfe2l2 gene), the main regulator of the antioxidant response, in muscle cell biology. We used (i) an immortalized murine myoblast cell line (C2C12) with stable overexpression of NRF2 and (ii) primary mSCs isolated from wild-type and Nfe2l2 (transcriptionally)-deficient mice (Nfe2l2tKO). NRF2 promoted myoblast proliferation and viability under oxidative stress conditions and decreased the production of reactive oxygen species. Furthermore, NRF2 overexpression inhibited C2C12 cell differentiation by down-regulating the expression of myogenic regulatory factors (MRFs) and muscle-specific microRNAs. We also showed that NRF2 is indispensable for the viability of mSCs since the lack of its transcriptional activity caused high mortality of cells cultured in vitro under normoxic conditions. Concomitantly, Nfe2l2tKO mSCs grown and differentiated under hypoxic conditions were viable and much more differentiated compared to cells isolated from wild-type mice. Taken together, NRF2 significantly influences the properties of myoblasts and muscle satellite cells. This effect might be modulated by the muscle microenvironment.
Collapse
|
11
|
Thomas NT, Confides AL, Fry CS, Dupont-Versteegden EE. Satellite cell depletion does not affect diaphragm adaptations to hypoxia. J Appl Physiol (1985) 2022; 133:637-646. [PMID: 35861521 PMCID: PMC9448290 DOI: 10.1152/japplphysiol.00083.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 01/25/2023] Open
Abstract
The diaphragm is the main skeletal muscle responsible for inspiration and is susceptible to age-associated decline in function and morphology. Satellite cells in diaphragm fuse into unperturbed muscle fibers throughout life, yet their role in adaptations to hypoxia in diaphragm is unknown. Given their continual fusion, we hypothesize that satellite cell depletion will negatively impact adaptations to hypoxia in the diaphragm, particularly with aging. We used the Pax7CreER/CreER:R26RDTA/DTA genetic mouse model of inducible satellite cell depletion to investigate diaphragm responses to hypoxia in adult (6 mo) and aged (22 mo) male mice. The mice were subjected to normobaric hypoxia at 10% [Formula: see text] or normoxia for 4 wk. We showed that satellite cell depletion had no effect on diaphragm muscle fiber cross-sectional area, fiber-type distribution, myonuclear density, or regulation of extracellular matrix in either adult or aged mice. Furthermore, we showed lower muscle fiber cross-sectional area with hypoxia and age (main effects), while extracellular matrix content was higher and satellite cell abundance was lower with age (main effect) in diaphragm. Lastly, a greater number of Pax3-mRNA+ cells was observed in diaphragm muscle of satellite cell-depleted mice independent of hypoxia (main effect), potentially as a compensatory mechanism for the loss of satellite cells. We conclude that satellite cells are not required for diaphragm muscle adaptations to hypoxia in either adult or aged mice.NEW & NOTEWORTHY Satellite cells show consistent fusion into diaphragm muscle fibers throughout life, suggesting a critical role in maintaining homeostasis. Here, we report identical diaphragm adaptations to hypoxia with and without satellite cells in adult and aged mice. In addition, we propose that the higher number of Pax3-positive cells in satellite cell-depleted diaphragm muscle acts as a compensatory mechanism.
Collapse
Affiliation(s)
- Nicholas T Thomas
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, Kentucky
| | - Amy L Confides
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky
| | - Christopher S Fry
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Athletic Training and Clinical Nutrition, University of Kentucky, Lexington, Kentucky
| | - Esther E Dupont-Versteegden
- Center for Muscle Biology, University of Kentucky, Lexington, Kentucky
- Department of Physical Therapy, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
12
|
Divvela SSK, Offei EB, Suerland F, Revuelta García D, Kwiatkowski J, Balakrishnan-Renuka A, Bohne P, Böing M, Morosan-Puopolo G, Mark MD, Brand-Saberi B. Atonal homolog 8/Math6 regulates differentiation and maintenance of skeletal muscle. Front Cell Dev Biol 2022; 10:950414. [PMID: 36060799 PMCID: PMC9438786 DOI: 10.3389/fcell.2022.950414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/06/2022] [Indexed: 12/16/2022] Open
Abstract
Atonal Homolog 8 (Atoh8) belongs to a large superfamily of transcriptional regulators called basic helix-loop-helix (bHLH) transcription factors. Atoh8 (murine homolog “Math6”) has been shown to be involved in organogenesis during murine embryonic development. We have previously identified the expression of Atoh8 during skeletal myogenesis in chicken where we described its involvement in hypaxial myotome formation suggesting a regulatory role of Atoh8 in skeletal muscle development. Within the current study, we analyzed the effect of the loss of function of Atoh8 in murine primary myoblasts and during differentiation of pluripotent stem cells into myotubes, and the effect of its gain of function in C2C12 cells. Based on the observed results, we conclude that Atoh8 regulates myoblast proliferation via modulating myostatin signaling. Further, our data revealed a reduced muscle mass, strength and fiber size with significant changes to the muscle fiber type suggesting atrophy in skeletal muscle of Atoh8 mutants. We further report that Atoh8 knockout mice suffer from a condition similar to ambient hypoxia which may be the primary cause of the phenotype. Altogether, this study shows the significance of Atoh8 not only in myogenesis but also in the maintenance of skeletal muscle.
Collapse
Affiliation(s)
| | - Eric Bekoe Offei
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- University of Ghana, School of Veterinary Medicine, Legon, Ghana
| | - Florian Suerland
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - David Revuelta García
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Julia Kwiatkowski
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Ajeesh Balakrishnan-Renuka
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Pauline Bohne
- Department of Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Marion Böing
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Gabriela Morosan-Puopolo
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Melanie D. Mark
- Department of Behavioral Neuroscience, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
- *Correspondence: Beate Brand-Saberi,
| |
Collapse
|
13
|
Ke D, Kengla C, Lee SJ, Yoo JJ, Zhu X, Murphy SV. Release Kinetics and In Vitro Characterization of Sodium Percarbonate and Calcium Peroxide to Oxygenate Bioprinted Tissue Models. Int J Mol Sci 2022; 23:ijms23126842. [PMID: 35743287 PMCID: PMC9224261 DOI: 10.3390/ijms23126842] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 01/27/2023] Open
Abstract
Oxygen-generating materials have been used in several tissue engineering applications; however, their application as in situ oxygen supply within bioprinted constructs has not been deeply studied. In this study, two oxygen-generating materials, sodium percarbonate (SPO) and calcium peroxide (CPO), were studied for their oxygen release kinetics under a 0.1% O2 condition. In addition, a novel cell-culture-insert setup was used to evaluate the effects of SPO and CPO on the viability of skeletal muscle cells under the same hypoxic condition. Results showed that SPO had a burst oxygen release, while CPO had a more stable oxygen release than SPO. Both SPO and CPO reduced cell viability when used alone. The addition of catalase in SPO and CPO increased the oxygen release rate, as well as improving the viability of skeletal muscle cells; however, CPO still showed cytotoxicity with catalase. Additionally, the utilization of 1 mg/mL SPO and 20 U catalase in a hydrogel for bioprinting significantly enhanced the cell viability under the hypoxic condition. Moreover, bioprinted muscle constructs could further differentiate into elongated myotubes when transferring back to the normoxic condition. This work provides an excellent in vitro model to test oxygen-generating materials and further discover their applications in bioprinting, where they represent promising avenues to overcome the challenge of oxygen shortage in bioprinted constructs before their complete vascularization.
Collapse
Affiliation(s)
- Dongxu Ke
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.K.); (C.K.); (S.J.L.); (J.J.Y.)
| | - Carlos Kengla
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.K.); (C.K.); (S.J.L.); (J.J.Y.)
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.K.); (C.K.); (S.J.L.); (J.J.Y.)
| | - James J. Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.K.); (C.K.); (S.J.L.); (J.J.Y.)
| | - Xuesong Zhu
- Department of Orthopedics, First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Correspondence: (X.Z.); (S.V.M.)
| | - Sean Vincent Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (D.K.); (C.K.); (S.J.L.); (J.J.Y.)
- Correspondence: (X.Z.); (S.V.M.)
| |
Collapse
|
14
|
Pircher T, Wackerhage H, Akova E, Böcker W, Aszodi A, Saller MM. Fusion of Normoxic- and Hypoxic-Preconditioned Myoblasts Leads to Increased Hypertrophy. Cells 2022; 11:cells11061059. [PMID: 35326510 PMCID: PMC8947054 DOI: 10.3390/cells11061059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/14/2022] [Accepted: 03/19/2022] [Indexed: 12/20/2022] Open
Abstract
Injuries, high altitude, and endurance exercise lead to hypoxic conditions in skeletal muscle and sometimes to hypoxia-induced local tissue damage. Thus, regenerative myoblasts/satellite cells are exposed to different levels and durations of partial oxygen pressure depending on the spatial distance from the blood vessels. To date, it is unclear how hypoxia affects myoblasts proliferation, differentiation, and particularly fusion with normoxic myoblasts. To study this, we investigated how 21% and 2% oxygen affects C2C12 myoblast morphology, proliferation, and myogenic differentiation and evaluated the fusion of normoxic- or hypoxic-preconditioned C2C12 cells in 21% or 2% oxygen in vitro. Out data show that the long-term hypoxic culture condition does not affect the proliferation of C2C12 cells but leads to rounder cells and reduced myotube formation when compared with myoblasts exposed to normoxia. However, when normoxic- and hypoxic-preconditioned myoblasts were differentiated together, the resultant myotubes were significantly larger than the control myotubes. Whole transcriptome sequencing analysis revealed several novel candidate genes that are differentially regulated during the differentiation under normoxia and hypoxia in mixed culture conditions and may thus be involved in the increase in myotube size. Taken together, oxygen-dependent adaption and interaction of myoblasts may represent a novel approach for the development of innovative therapeutic targets.
Collapse
Affiliation(s)
- Tamara Pircher
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Henning Wackerhage
- Faculty of Sport and Health Sciences, Technical University of Munich, Georg-Brauchle-Ring 60, 80992 Munich, Germany;
| | - Elif Akova
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Wolfgang Böcker
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Attila Aszodi
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
| | - Maximilian M. Saller
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), Ludwig-Maximilians-University (LMU), Fraunhoferstraße 20, 82152 Planegg-Martinsried, Germany; (T.P.); (E.A.); (W.B.); (A.A.)
- Correspondence: ; Tel.: +49-89-4400-55486
| |
Collapse
|
15
|
Elashry MI, Kinde M, Klymiuk MC, Eldaey A, Wenisch S, Arnhold S. The effect of hypoxia on myogenic differentiation and multipotency of the skeletal muscle-derived stem cells in mice. Stem Cell Res Ther 2022; 13:56. [PMID: 35123554 PMCID: PMC8817503 DOI: 10.1186/s13287-022-02730-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/20/2022] [Indexed: 01/01/2023] Open
Abstract
Abstract
Background
Skeletal muscle-derived stem cells (SC) have become a promising approach for investigating myogenic differentiation and optimizing tissue regeneration. Muscle regeneration is performed by SC, a self-renewal cell population underlying the basal lamina of muscle fibers. Here, we examined the impact of hypoxia condition on the regenerative capacity of SC either in their native microenvironment or via isolation in a monolayer culture using ectopic differentiation inductions. Furthermore, the effect of low oxygen tension on myogenic differentiation protocols of the myoblasts cell line C2C12 was examined.
Methods
Hind limb muscles of wild type mice were processed for both SC/fiber isolation and myoblast extraction using magnetic beads. SC were induced for myogenic, adipogenic and osteogenic commitments under normoxic (21% O2) and hypoxic (3% O2) conditions. SC proliferation and differentiation were evaluated using histological staining, immunohistochemistry, morphometric analysis and RT-qPCR. The data were statistically analyzed using ANOVA.
Results
The data revealed enhanced SC proliferation and motility following differentiation induction after 48 h under hypoxia. Following myogenic induction, the number of undifferentiated cells positive for Pax7 were increased at 72 h under hypoxia. Hypoxia upregulated MyoD and downregulated Myogenin expression at day-7 post-myogenic induction. Hypoxia promoted both SC adipogenesis and osteogenesis under respective induction as shown by using Oil Red O and Alizarin Red S staining. The expression of adipogenic markers; peroxisome proliferator activated receptor gamma (PPARγ) and fatty acid-binding protein 4 (FABP4) were upregulated under hypoxia up to day 14 compared to normoxic condition. Enhanced osteogenic differentiation was detected under hypoxic condition via upregulation of osteocalcin and osteopontin expression up to day 14 as well as, increased calcium deposition at day 21. Hypoxia exposure increases the number of adipocytes and the size of fat vacuoles per adipocyte compared to normoxic culture. Combining the differentiation medium with dexamethasone under hypoxia improves the efficiency of the myogenic differentiation protocol of C2C12 by increasing the length of the myotubes.
Conclusions
Hypoxia exposure increases cell resources for clinical applications and promotes SC multipotency and thus beneficial for tissue regeneration.
Collapse
|
16
|
Bensaid S, Fabre C, Pawlak-Chaouch M, Cieniewski-Bernard C. Oxygen supplementation to limit hypoxia-induced muscle atrophy in C2C12 myotubes: comparison with amino acid supplement and electrical stimulation. Cell Tissue Res 2022; 387:287-301. [PMID: 35001209 DOI: 10.1007/s00441-021-03492-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/21/2021] [Indexed: 11/28/2022]
Abstract
In skeletal muscle, chronic oxygen depletion induces a disturbance leading to muscle atrophy. Mechanical stress (physical exercise) and nutritional supplement therapy are commonly used against loss of muscle mass and undernutrition in hypoxia, while oxygenation therapy is preferentially used to counteract muscle fatigue and exercise intolerance. However, the impact of oxygenation on skeletal muscle cells remains poorly understood, in particular on signalling pathways regulating protein balance. Thus, we investigated the effects of each separated treatment (mechanical stress, nutritional supplementation and oxygenation therapy) on intracellular pathways involved in protein synthesis and degradation that are imbalanced in skeletal muscle cells atrophy resulting from hypoxia. Myotubes under hypoxia were treated by electrical stimulation, amino acids supplement or oxygenation period. Signalling pathways involved in protein synthesis (PI3K-Akt-mTOR) and degradation (FoxO1 and FoxO3a) were investigated, so as autophagy, ubiquitin-proteasome system and myotube morphology. Electrical stimulation and oxygenation treatment resulted in higher myotube diameter, myogenic fusion index and myotubes density until 48 h post-treatment compared to untreated hypoxic myotubes. Both treatments also induced inhibition of FoxO3a and decreased activity of ubiquitin-proteasome system; however, their impact on protein synthesis pathway was specific for each one. Indeed, electrical stimulation impacted upstream proteins to mTOR (i.e., Akt) while oxygenation treatment activated downstream targets of mTOR (i.e., 4E-BP1 and P70S6K). In contrast, amino acid supplementation had very few effects on myotube morphology nor on protein homeostasis. This study demonstrated that electrical stimulation or oxygenation period are two effective treatments to fight against hypoxia-induced muscle atrophy, acting through different molecular adaptations.
Collapse
Affiliation(s)
- Samir Bensaid
- Univ. Lille, Univ. Artois, Univ. Littoral Côte D'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000, Lille, France.,CHU Lille, Université de Lille, F-59000, Lille, France
| | - Claudine Fabre
- Univ. Lille, Univ. Artois, Univ. Littoral Côte D'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000, Lille, France
| | - Mehdi Pawlak-Chaouch
- Univ. Lille, Univ. Artois, Univ. Littoral Côte D'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000, Lille, France
| | - Caroline Cieniewski-Bernard
- Univ. Lille, Univ. Artois, Univ. Littoral Côte D'Opale, URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, F-59000, Lille, France.
| |
Collapse
|
17
|
Zhao L, Law NC, Gomez NA, Son J, Gao Y, Liu X, de Avila JM, Zhu M, Du M. Obesity Impairs Embryonic Myogenesis by Enhancing BMP Signaling within the Dermomyotome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102157. [PMID: 34647690 PMCID: PMC8596142 DOI: 10.1002/advs.202102157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/16/2021] [Indexed: 05/05/2023]
Abstract
Obesity during pregnancy leads to adverse health outcomes in offspring. However, the initial effects of maternal obesity (MO) on embryonic organogenesis have yet to be thoroughly examined. Using unbiased single-cell transcriptomic analyses (scRNA-seq), the effects of MO on the myogenic process is investigated in embryonic day 9.5 (E9.5) mouse embryos. The results suggest that MO induces systematic hypoxia, which is correlated with enhanced BMP signaling and impairs skeletal muscle differentiation within the dermomyotome (DM). The Notch-signaling effectors, HES1 and HEY1, which also act down-stream of BMP signaling, suppress myogenic differentiation through transcriptionally repressing the important myogenic regulator MEF2C. Moreover, the major hypoxia effector, HIF1A, enhances expression of HES1 and HEY1 and blocks myogenic differentiation in vitro. In summary, this data demonstrate that MO induces hypoxia and impairs myogenic differentiation by up-regulating BMP signaling within the DM, which may account for the disruptions of skeletal muscle development and function in progeny.
Collapse
Affiliation(s)
- Liang Zhao
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciencesand School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
| | - Nathan C. Law
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
- Center for Reproductive BiologyCollege of Veterinary MedicineWashington State UniversityPullmanWA99164USA
| | - Noe A. Gomez
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciencesand School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
| | - Junseok Son
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciencesand School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
| | - Yao Gao
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciencesand School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
| | - Xiangdong Liu
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciencesand School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
| | - Jeanene M. de Avila
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciencesand School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
| | - Mei‐Jun Zhu
- School of Food ScienceWashington State UniversityPullmanWA99164USA
| | - Min Du
- Nutrigenomics and Growth Biology LaboratoryDepartment of Animal Sciencesand School of Molecular BioscienceWashington State UniversityPullmanWA99164USA
- Department of Animal SciencesWashington State UniversityPullmanWA99164USA
| |
Collapse
|
18
|
Horiike M, Ogawa Y, Kawada S. Effects of hyperoxia and hypoxia on the proliferation of C2C12 myoblasts. Am J Physiol Regul Integr Comp Physiol 2021; 321:R572-R587. [PMID: 34431403 DOI: 10.1152/ajpregu.00269.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Hyperoxic conditions are known to accelerate skeletal muscle regeneration after injuries. In the early phase of regeneration, macrophages invade the injured area and subsequently secrete various growth factors, which regulate myoblast proliferation and differentiation. Although hyperoxic conditions accelerate muscle regeneration, it is unknown whether this effect is indirectly mediated by macrophages. Here, using C2C12 cells, we show that not only hyperoxia but also hypoxia enhance myoblast proliferation directly, without accelerating differentiation into myotubes. Under hyperoxic conditions (95% O2 + 5% CO2), the cell membrane was damaged because of lipid oxidization, and a disrupted cytoskeletal structure, resulting in suppressed cell proliferation. However, a culture medium containing vitamin C (VC), an antioxidant, prevented this lipid oxidization and cytoskeletal disruption, resulting in enhanced proliferation in response to hyperoxia exposure of ≤4 h/day. In contrast, exposure to hypoxic conditions (95% N2 + 5% CO2) for ≤8 h/day enhanced cell proliferation. Hyperoxia did not promote cell differentiation into myotubes, regardless of whether the culture medium contained VC. Similarly, hypoxia did not accelerate cell differentiation. These results suggest that regardless of hyperoxia or hypoxia, changes in oxygen tension can enhance cell proliferation directly, but do not influence differentiation efficiency in C2C12 cells. Moreover, excess oxidative stress abrogated the enhancement of myoblast proliferation induced by hyperoxia. This research will contribute to basic data for applying the effects of hyperoxia or hypoxia to muscle regeneration therapy.
Collapse
Affiliation(s)
- Misa Horiike
- Department of Sport and Medical Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Yoshiko Ogawa
- Department of Sport and Medical Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| | - Shigeo Kawada
- Department of Sport and Medical Science, Faculty of Medical Technology, Teikyo University, Tokyo, Japan
| |
Collapse
|
19
|
Rozance PJ, Wesolowski SR, Jonker SS, Brown LD. Anemic hypoxemia reduces myoblast proliferation and muscle growth in late-gestation fetal sheep. Am J Physiol Regul Integr Comp Physiol 2021; 321:R352-R363. [PMID: 34287074 PMCID: PMC8530759 DOI: 10.1152/ajpregu.00342.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fetal skeletal muscle growth requires myoblast proliferation, differentiation, and fusion into myofibers in addition to protein accretion for fiber hypertrophy. Oxygen is an important regulator of this process. Therefore, we hypothesized that fetal anemic hypoxemia would inhibit skeletal muscle growth. Studies were performed in late-gestation fetal sheep that were bled to anemic and therefore hypoxemic conditions beginning at ∼125 days of gestation (term = 148 days) for 9 ± 0 days (n = 19) and compared with control fetuses (n = 16). A metabolic study was performed on gestational day ∼134 to measure fetal protein kinetic rates. Myoblast proliferation and myofiber area were determined in biceps femoris (BF), tibialis anterior (TA), and flexor digitorum superficialis (FDS) muscles. mRNA expression of muscle regulatory factors was determined in BF. Fetal arterial hematocrit and oxygen content were 28% and 52% lower, respectively, in anemic fetuses. Fetal weight and whole body protein synthesis, breakdown, and accretion rates were not different between groups. Hindlimb length, however, was 7% shorter in anemic fetuses. TA and FDS muscles weighed less, and FDS myofiber area was smaller in anemic fetuses compared with controls. The percentage of Pax7+ myoblasts that expressed Ki67 was lower in BF and tended to be lower in FDS from anemic fetuses indicating reduced myoblast proliferation. There was less MYOD and MYF6 mRNA expression in anemic versus control BF consistent with reduced myoblast differentiation. These results indicate that fetal anemic hypoxemia reduced muscle growth. We speculate that fetal muscle growth may be improved by strategies that increase oxygen availability.
Collapse
Affiliation(s)
- Paul J. Rozance
- 1Department of Pediatrics, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Stephanie R. Wesolowski
- 1Department of Pediatrics, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sonnet S. Jonker
- 2Center for Developmental Health, Knight Cardiovascular Institute,
Oregon Health & Science University, Portland, Oregon
| | - Laura D. Brown
- 1Department of Pediatrics, Perinatal Research Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
20
|
Nguyen TH, Conotte S, Belayew A, Declèves AE, Legrand A, Tassin A. Hypoxia and Hypoxia-Inducible Factor Signaling in Muscular Dystrophies: Cause and Consequences. Int J Mol Sci 2021; 22:7220. [PMID: 34281273 PMCID: PMC8269128 DOI: 10.3390/ijms22137220] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/29/2022] Open
Abstract
Muscular dystrophies (MDs) are a group of inherited degenerative muscle disorders characterized by a progressive skeletal muscle wasting. Respiratory impairments and subsequent hypoxemia are encountered in a significant subgroup of patients in almost all MD forms. In response to hypoxic stress, compensatory mechanisms are activated especially through Hypoxia-Inducible Factor 1 α (HIF-1α). In healthy muscle, hypoxia and HIF-1α activation are known to affect oxidative stress balance and metabolism. Recent evidence has also highlighted HIF-1α as a regulator of myogenesis and satellite cell function. However, the impact of HIF-1α pathway modifications in MDs remains to be investigated. Multifactorial pathological mechanisms could lead to HIF-1α activation in patient skeletal muscles. In addition to the genetic defect per se, respiratory failure or blood vessel alterations could modify hypoxia response pathways. Here, we will discuss the current knowledge about the hypoxia response pathway alterations in MDs and address whether such changes could influence MD pathophysiology.
Collapse
Affiliation(s)
- Thuy-Hang Nguyen
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Stephanie Conotte
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Alexandra Belayew
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Anne-Emilie Declèves
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium;
| | - Alexandre Legrand
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| | - Alexandra Tassin
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000 Mons, Belgium; (T.-H.N.); (S.C.); (A.B.); (A.L.)
| |
Collapse
|
21
|
Pircher T, Wackerhage H, Aszodi A, Kammerlander C, Böcker W, Saller MM. Hypoxic Signaling in Skeletal Muscle Maintenance and Regeneration: A Systematic Review. Front Physiol 2021; 12:684899. [PMID: 34248671 PMCID: PMC8260947 DOI: 10.3389/fphys.2021.684899] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/26/2021] [Indexed: 12/26/2022] Open
Abstract
In skeletal muscle tissue, oxygen (O2) plays a pivotal role in both metabolism and the regulation of several intercellular pathways, which can modify proliferation, differentiation and survival of cells within the myogenic lineage. The concentration of oxygen in muscle tissue is reduced during embryogenesis and pathological conditions. Myogenic progenitor cells, namely satellite cells, are necessary for muscular regeneration in adults and are localized in a hypoxic microenvironment under the basal lamina, suggesting that the O2 level could affect their function. This review presents the effects of reduced oxygen levels (hypoxia) on satellite cell survival, myoblast regeneration and differentiation in vertebrates. Further investigations and understanding of the pathways involved in adult muscle regeneration during hypoxic conditions are maybe clinically relevant to seek for novel drug treatments for patients with severe muscle damage. We especially outlined the effect of hypoxia-inducible factor 1-alpha (HIF1A), the most studied transcriptional regulator of cellular and developmental response to hypoxia, whose investigation has recently been awarded with the Nobel price.
Collapse
Affiliation(s)
- Tamara Pircher
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Henning Wackerhage
- Faculty of Sport and Health Sciences, Technical University of Munich, Munich, Germany
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Christian Kammerlander
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Wolfgang Böcker
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| | - Maximilian Michael Saller
- Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstructive Surgery, Munich University Hospital, Munich, Germany
| |
Collapse
|
22
|
van Doorslaer de Ten Ryen S, Francaux M, Deldicque L. Regulation of satellite cells by exercise in hypoxic conditions: a narrative review. Eur J Appl Physiol 2021; 121:1531-1542. [PMID: 33745023 DOI: 10.1007/s00421-021-04641-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/10/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE To investigate in vivo the adaptations of satellite cell induced by exercise performed in acute or chronic hypoxic conditions and their contribution to muscle remodeling and hypertrophy. METHODS Search terms related to exercise, hypoxia and satellite cells were entered on Embase, PubMed and Scopus. Studies were selected for their relevance in terms of regulation of satellite cells by in vivo exercise and muscle contraction in hypoxic conditions. RESULTS Satellite cell activation and proliferation seem to be enabled after acute hypoxic exercise via regulations induced by myogenic regulatory factors. Several studies reported also a role of the inflammatory pathway nuclear factor-kappa B and angiogenic factors such as vascular endothelial growth factor, both known to upregulate myogenesis. By stimulating angiogenesis, repeated exercise performed in acute hypoxia might contribute to satellite cell activation. Contrary to such exercise conditions, chronic exposure to hypoxia downregulates myogenesis despite the maintenance of physical activity. This impaired myogenesis might be induced by excessive oxidative stress and proteolysis. CONCLUSION In vivo studies suggest that, in comparison to exercise or hypoxia alone, exercise performed in a hypoxic environment, may improve or impair muscle remodeling induced by contractile activity depending upon the duration of hypoxia. Satellite cells seem to be major actors in these dichotomous adaptations. Further research on the role of angiogenesis, types of contraction and autophagy is needed for a better understanding of their respective role in hypoxic exercise-induced modulations of satellite cell activity in human.
Collapse
Affiliation(s)
- Sophie van Doorslaer de Ten Ryen
- Institute of Neuroscience, Université Catholique de Louvain, Place Pierre de Coubertin, 1 L08.10.01, 1348, Louvain-la-Neuve, Belgium
| | - Marc Francaux
- Institute of Neuroscience, Université Catholique de Louvain, Place Pierre de Coubertin, 1 L08.10.01, 1348, Louvain-la-Neuve, Belgium
| | - Louise Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Place Pierre de Coubertin, 1 L08.10.01, 1348, Louvain-la-Neuve, Belgium.
| |
Collapse
|
23
|
Krist B, Podkalicka P, Mucha O, Mendel M, Sępioł A, Rusiecka OM, Józefczuk E, Bukowska-Strakova K, Grochot-Przęczek A, Tomczyk M, Klóska D, Giacca M, Maga P, Niżankowski R, Józkowicz A, Łoboda A, Dulak J, Florczyk-Soluch U. miR-378a influences vascularization in skeletal muscles. Cardiovasc Res 2021; 116:1386-1397. [PMID: 31504257 DOI: 10.1093/cvr/cvz236] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/28/2019] [Accepted: 08/22/2019] [Indexed: 12/14/2022] Open
Abstract
AIMS MicroRNA-378a, highly expressed in skeletal muscles, was demonstrated to affect myoblasts differentiation and to promote tumour angiogenesis. We hypothesized that miR-378a could play a pro-angiogenic role in skeletal muscle and may be involved in regeneration after ischaemic injury in mice. METHODS AND RESULTS Silencing of miR-378a in murine C2C12 myoblasts did not affect differentiation but impaired their secretory angiogenic potential towards endothelial cells. miR-378a knockout (miR-378a-/-) in mice resulted in a decreased number of CD31-positive blood vessels and arterioles in gastrocnemius muscle. In addition, diminished endothelial sprouting from miR-378a-/- aortic rings was shown. Interestingly, although fibroblast growth factor 1 (Fgf1) expression was decreased in miR-378a-/- muscles, this growth factor did not mediate the angiogenic effects exerted by miR-378a. In vivo, miR-378a knockout did not affect the revascularization of the ischaemic muscles in both normo- and hyperglycaemic mice subjected to femoral artery ligation (FAL). No difference in regenerating muscle fibres was detected between miR-378a-/- and miR-378+/+ mice. miR-378a expression temporarily declined in ischaemic skeletal muscles of miR-378+/+ mice already on Day 3 after FAL. At the same time, in the plasma, the level of miR-378a-3p was enhanced. Similar elevation of miR-378a-3p was reported in the plasma of patients with intermittent claudication in comparison to healthy donors. Local adeno-associated viral vectors-based miR-378a overexpression was enough to improve the revascularization of the ischaemic limb of wild-type mice on Day 7 after FAL, what was not reported after systemic delivery of vectors. In addition, the number of infiltrating CD45+ cells and macrophages (CD45+ CD11b+ F4/80+ Ly6G-) was higher in the ischaemic muscles of miR-378a-/- mice, suggesting an anti-inflammatory action of miR-378a. CONCLUSIONS Data indicate miR-378a role in the pro-angiogenic effect of myoblasts and vascularization of skeletal muscle. After the ischaemic insult, the anti-angiogenic effect of miR-378a deficiency might be compensated by enhanced inflammation.
Collapse
Affiliation(s)
- Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Olga Mucha
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Mateusz Mendel
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Aleksandra Sępioł
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Olga Martyna Rusiecka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Ewelina Józefczuk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.,Department of Clinical Immunology and Transplantology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Grochot-Przęczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Mateusz Tomczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Damian Klóska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, London, UK
| | - Paweł Maga
- Department of Angiology, Faculty of Medicine, Jagiellonian University, Krakow, Poland
| | - Rafał Niżankowski
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, London, UK
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland.,Kardio-Med Silesia, Zabrze, Poland
| | - Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| |
Collapse
|
24
|
Choi KH, Yoon JW, Kim M, Lee HJ, Jeong J, Ryu M, Jo C, Lee CK. Muscle stem cell isolation and in vitro culture for meat production: A methodological review. Compr Rev Food Sci Food Saf 2021; 20:429-457. [PMID: 33443788 DOI: 10.1111/1541-4337.12661] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Cultured muscle tissue-based protein products, also known as cultured meat, are produced through in vitro myogenesis involving muscle stem cell culture and differentiation, and mature muscle cell processing for flavor and texture. This review focuses on the in vitro myogenesis for cultured meat production. The muscle stem cell-based in vitro muscle tissue production consists of a sequential process: (1) muscle sampling for stem cell collection, (2) muscle tissue dissociation and muscle stem cell isolation, (3) primary cell culture, (4) upscaled cell culture, (5) muscle differentiation and maturation, and (6) muscle tissue harvest. Although muscle stem cell research is a well-established field, the majority of these steps remain to be underoptimized to enable the in vitro creation of edible muscle-derived meat products. The profound understanding of the process would help not only cultured meat production but also business sectors that have been seeking new biomaterials for the food industry. In this review, we discuss comprehensively and in detail each step of cutting-edge methods for cultured meat production. This would be meaningful for both academia and industry to prepare for the new era of cellular agriculture.
Collapse
Affiliation(s)
- Kwang-Hwan Choi
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Ji Won Yoon
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Minsu Kim
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Hyun Jung Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Jinsol Jeong
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Minkyung Ryu
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Cheorun Jo
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
| |
Collapse
|
25
|
Cirillo F, Resmini G, Angelino E, Ferrara M, Tarantino A, Piccoli M, Rota P, Ghiroldi A, Monasky MM, Ciconte G, Pappone C, Graziani A, Anastasia L. HIF-1α Directly Controls WNT7A Expression During Myogenesis. Front Cell Dev Biol 2020; 8:593508. [PMID: 33262987 PMCID: PMC7686515 DOI: 10.3389/fcell.2020.593508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/20/2020] [Indexed: 11/13/2022] Open
Abstract
Herein we unveil that Hypoxia-inducible factor-1α (HIF-1α) directly regulates WNT7A expression during myogenesis. In fact, chromatin immunoprecipitation (ChiP) and site-directed mutagenesis experiments revealed two distinct hypoxia response elements (HREs) that are specific HIF-1α binding sites on the WNT7A promoter. Remarkably, a pharmacological activation of HIF-1α induced WNT7A expression and enhanced muscle differentiation. On the other hand, silencing of WNT7A using CRISPR/Cas9 genome editing blocked the effects of HIF-1α activation on myogenesis. Finally, treatment with prolyl hydroxylases (PHDs) inhibitors improved muscle regeneration in vitro and in vivo in a cardiotoxin (CTX)-induced muscle injury mouse model, paving the way for further studies to test its efficacy on acute and chronic muscular pathologies.
Collapse
Affiliation(s)
- Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Giulia Resmini
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Elia Angelino
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Michele Ferrara
- Division of Genetics and Cell Biology, Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Adriana Tarantino
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | - Paola Rota
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy
| | | | - Giuseppe Ciconte
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, Milan, Italy.,Vita-Salute San Raffaele University, Faculty of Medicine, Milan, Italy
| | - Andrea Graziani
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, San Donato Milanese, Italy.,Vita-Salute San Raffaele University, Faculty of Medicine, Milan, Italy
| |
Collapse
|
26
|
Gosker HR, Langen RC, Simons SO. Role of acute exacerbations in skeletal muscle impairment in COPD. Expert Rev Respir Med 2020; 15:103-115. [PMID: 33131350 DOI: 10.1080/17476348.2021.1843429] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Introduction: Muscle impairments are prevalent in COPD and have adverse clinical implications in terms of physical performance capacity, disease burden, quality of life and even mortality. During acute exacerbations of COPD (AECOPDs) the respiratory symptoms worsen and this might also apply to the muscle impairments. Areas covered: This report includes a review of both clinical and pre-clinical peer-reviewed literature of the past 20 years found in PubMed providing a comprehensive view on the role of AECOPD in muscle dysfunction in COPD, the putative underlying mechanisms and the treatment perspectives. Expert opinion: The contribution of AECOPD and its recurrent nature to muscle impairment in COPD cannot be ignored and can be attributed to the acutely intensifying and converging disease-related drivers of muscle deterioration, in particular disuse, systemic inflammation and corticosteroid treatment. The search for novel treatment options should focus on the AECOPD-enhanced drivers of muscle dysfunction as well as on the underlying, mainly catabolic, mechanisms. Considering the impact of AECOPD on muscle function, and that of muscle impairment on the recurrence of exacerbations, counteracting muscle deterioration in AECOPD provides an unprecedented therapeutic opportunity.
Collapse
Affiliation(s)
- Harry R Gosker
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Department of Respiratory Medicine , Maastricht, The Netherlands
| | - Ramon C Langen
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Department of Respiratory Medicine , Maastricht, The Netherlands
| | - Sami O Simons
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Department of Respiratory Medicine , Maastricht, The Netherlands
| |
Collapse
|
27
|
Chopra A, Cho WC, Willmore WG, Biggar KK. Hypoxia-Inducible Lysine Methyltransferases: G9a and GLP Hypoxic Regulation, Non-histone Substrate Modification, and Pathological Relevance. Front Genet 2020; 11:579636. [PMID: 33088284 PMCID: PMC7495024 DOI: 10.3389/fgene.2020.579636] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/13/2020] [Indexed: 12/29/2022] Open
Abstract
Oxygen sensing is inherent among most animal lifeforms and is critical for organism survival. Oxygen sensing mechanisms collectively trigger cellular and physiological responses that enable adaption to a reduction in ideal oxygen levels. The major mechanism by which oxygen-responsive changes in the transcriptome occur are mediated through the hypoxia-inducible factor (HIF) pathway. Upon reduced oxygen conditions, HIF activates hypoxia-responsive gene expression programs. However, under normal oxygen conditions, the activity of HIF is regularly suppressed by cellular oxygen sensors; prolyl-4 and asparaginyl hydroxylases. Recently, these oxygen sensors have also been found to suppress the function of two lysine methyltransferases, G9a and G9a-like protein (GLP). In this manner, the methyltransferase activity of G9a and GLP are hypoxia-inducible and thus present a new avenue of low-oxygen signaling. Furthermore, G9a and GLP elicit lysine methylation on a wide variety of non-histone proteins, many of which are known to be regulated by hypoxia. In this article we aim to review the effects of oxygen on G9a and GLP function, non-histone methylation events inflicted by these methyltransferases, and the clinical relevance of these enzymes in cancer.
Collapse
Affiliation(s)
- Anand Chopra
- Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, China
| | - William G. Willmore
- Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Kyle K. Biggar
- Institute of Biochemistry, Carleton University, Ottawa, ON, Canada
- Department of Biology, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
28
|
Feng S, Ma J, Long K, Zhang J, Qiu W, Li Y, Jin L, Wang X, Jiang A, Liu L, Xiao W, Li X, Tang Q, Li M. Comparative microRNA Transcriptomes in Domestic Goats Reveal Acclimatization to High Altitude. Front Genet 2020; 11:809. [PMID: 32849809 PMCID: PMC7411263 DOI: 10.3389/fgene.2020.00809] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 07/06/2020] [Indexed: 01/30/2023] Open
Abstract
High-altitude acclimatization is a representative example of vertebrates' acclimatization to harsh and extreme environments. Previous studies reported sufficient evidence for a molecular genetic basis of high-altitude acclimatization, and genomic patterns of genetic variation among populations and species have been widely elucidated in recent years. However, understanding of the miRNA role in high-altitude acclimatization have lagged behind, especially in non-model species. To investigate miRNA expression alterations of goats that were induced by high-altitude stress, we performed comparative miRNA transcriptome analysis on six hypoxia-sensitive tissues (heart, kidney, liver, lung, skeletal muscle, and spleen) in two goat populations from distinct altitudes (600 and 3000 m). We obtained the expression value of 1391 mature miRNAs and identified 138 differentially expressed (DE) miRNAs between high and low altitudes. Combined with tissue specificity analysis, we illustrated alterations of expression levels among altitudes and tissues, and found that there were coexisting tissue-specific and -conserved mechanisms for hypoxia acclimatization. Notably, the interplay between DE miRNA and DE target genes strongly indicated post-transcriptional regulation in the hypoxia inducible factor 1, insulin, and p53 signaling pathways, which might play significant roles in high-altitude acclimatization in domestic goats. It's also worth noting that we experimentally confirmed miR-106a-5p to have a negative regulation effect on angiogenesis by directly targeting FLT-1. These results provide insight into the complicated miRNA expression patterns and regulatory mechanisms of high-altitude acclimatization in domestic goats.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Qianzi Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
29
|
Wagatsuma A, Arakawa M, Matsumoto H, Matsuda R, Hoshino T, Mabuchi K. Cobalt chloride, a chemical hypoxia-mimicking agent, suppresses myoblast differentiation by downregulating myogenin expression. Mol Cell Biochem 2020; 470:199-214. [PMID: 32451753 DOI: 10.1007/s11010-020-03762-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 05/16/2020] [Indexed: 12/17/2022]
Abstract
Cobalt chloride can create hypoxia-like state in vitro (referred to as chemical hypoxia). Several studies have suggested that chemical hypoxia may cause deleterious effects on myogenesis. The intrinsic underlying mechanisms of myoblast differentiation, however, are not fully understood. Here, we show that cobalt chloride strongly suppresses myoblast differentiation in a dose-dependent manner. The impaired myoblast differentiation is accompanied by downregulation of myogenic regulatory factor myogenin. Under chemical hypoxia, myogenin stability is decreased at mRNA and protein levels. A muscle-specific E3 ubiquitin ligase MAFbx, which can target myogenin protein for proteasomal degradation, is upregulated along with changes in Akt/Foxo and AMPK/Foxo signaling pathways. A proteasome inhibitor completely prevents cobalt chloride-mediated decrease in myogenin protein. These results suggest that cobalt chloride might modulate myogenin expression at post-transcriptional and post-translational levels, resulting in the failure of the myoblasts to differentiate into myotubes.
Collapse
Affiliation(s)
- Akira Wagatsuma
- Department of Information Physics and Computing, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan.
- Department of Communication, Tokyo Women's Christian University, Tokyo, Japan.
| | - Masayuki Arakawa
- Institute of Microbial Chemistry, Biology Division, Laboratory of Virology, Tokyo, Japan
| | - Hanano Matsumoto
- Department of Food and Health Science, Faculty of Human Life Sciences, Jissen Women's University, Tokyo, Japan
| | - Ryoichi Matsuda
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, Japan
| | - Takayuki Hoshino
- Department of Information Physics and Computing, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kunihiko Mabuchi
- Department of Information Physics and Computing, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
Prolyl hydroxylase domain 2 reduction enhances skeletal muscle tissue regeneration after soft tissue trauma in mice. PLoS One 2020; 15:e0233261. [PMID: 32413092 PMCID: PMC7228053 DOI: 10.1371/journal.pone.0233261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
The transcription factor Hypoxia-inducible factor 1 (HIF-1) plays a pivotal role in tissue regeneration. HIF-1 is negatively controlled by O2-dependent prolyl hydroxylases with a predominant role of prolyl hydroxylase 2 isoform (Phd2). Transgenic mice, hypomorphic for this isoform, accumulate more HIF-1 under normoxic conditions. Using these mice, we investigated the influence of Phd2 and HIF-1 on the regenerative capability of skeletal muscle tissue after myotrauma. Phd2-hypomorphic and wild type mice (on C57Bl/6 background) were grouped with regeneration times from 6 to 168 hours after closed mechanic muscle trauma to the hind limb. Tissue samples were analysed by immuno-staining and real-time PCR. Bone marrow derived macrophages of wild type and Phd2-hypomorphic mice were isolated and analysed via flow cytometry and quantitative real-time PCR. Phd2 reduction led to a higher regenerative capability due to enhanced activation of myogenic factors accompanied by induction of genes responsible for glucose and lactate metabolism in Phd2-hypomorphic mice. Macrophage infiltration into the trauma areas in hypomorphic mice started earlier and was more pronounced compared to wild type mice. Phd2-hypomorphic mice also showed higher numbers of macrophages in areas with sustained trauma 72 hours after myotrauma application. In conclusion, we postulate that the HIF-1 pathway is activated secondary to a Phd2 reduction which may lead to i) higher activation of myogenic factors, ii) increased number of positive stem cell proliferation markers, and iii) accelerated macrophage recruitment to areas of trauma, resulting in faster muscle tissue regeneration after myotrauma. With the current development of prolyl hydroxylase domain inhibitors, our findings point towards a potential clinical benefit after myotrauma.
Collapse
|
31
|
Zhang W, Yu L, Han X, Pan J, Deng J, Zhu L, Lu Y, Huang W, Liu S, Li Q, Liu Y. The secretome of human dental pulp stem cells protects myoblasts from hypoxia‑induced injury via the Wnt/β‑catenin pathway. Int J Mol Med 2020; 45:1501-1513. [PMID: 32323739 PMCID: PMC7138287 DOI: 10.3892/ijmm.2020.4525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022] Open
Abstract
Human dental pulp stem cells (hDPSCs) present several advantages, including their ability to be non-invasively harvested without ethical concern. The secretome of hDPSCs can promote the functional recovery of various tissue injuries. However, the protective effects on hypoxia-induced skeletal muscle injury remain to be explored. The present study demonstrated that C2C12 myoblast coculture with hDPSCs attenuated CoCl2-induced hypoxic injury compared with C2C12 alone. The hDPSC secretome increased cell viability and differentiation and decreased G2/M cell cycle arrest under hypoxic conditions. These results were further verified using hDPSC-conditioned medium (hDPSC-CM). The present data revealed that the protective effects of hDPSC-CM depend on the concentration ratio of the CM. In terms of the underlying molecular mechanism, hDPSC-CM activated the Wnt/β-catenin pathway, which increased the protein levels of Wnt1, phosphorylated-glycogen synthase kinase-3β and β-catenin and the mRNA levels of Wnt target genes. By contrast, an inhibitor (XAV939) of Wnt/β-catenin diminished the protective effects of hDPSC-CM. Taken together, the findings of the present study demonstrated that the hDPSC secretome alleviated the hypoxia-induced myoblast injury potentially through regulating the Wnt/β-catenin pathway. These findings may provide new insight into a therapeutic alternative using the hDPSC secretome in skeletal muscle hypoxia-related diseases.
Collapse
Affiliation(s)
- Weihua Zhang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Liming Yu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Xinxin Han
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Jie Pan
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Jiajia Deng
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Luying Zhu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Yun Lu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Wei Huang
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Shangfeng Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Qiang Li
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200001, P.R. China
| |
Collapse
|
32
|
Tian JJ, Fu B, Yu EM, Li YP, Xia Y, Li ZF, Zhang K, Gong WB, Yu DG, Wang GJ, Xie J. Feeding Faba Beans ( Vicia faba L.) Reduces Myocyte Metabolic Activity in Grass Carp ( Ctenopharyngodon idellus). Front Physiol 2020; 11:391. [PMID: 32395106 PMCID: PMC7197471 DOI: 10.3389/fphys.2020.00391] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 04/01/2020] [Indexed: 12/30/2022] Open
Abstract
In this study, we aimed to explore the effects of faba bean (Vicia faba L.) on the energy metabolism of grass carp (Ctenopharyngodon idellus). A total of 180 fish (∼2900 g) were randomly assigned to six tanks (2.5 × 2.5 × 1.2 m; 30 individuals per tank) and fed either faba bean (Vicia faba L.) or a commercial diet for 120 days (3% body weight, twice per day). The results showed that faba bean-fed grass carp (FBFG) had significantly lower growth and higher fat accumulation in the mesenteric adipose tissue and hepatopancreas than commercial diet-fed grass carp (CDFG). Compared with CDFG, FBFG exhibited no significant difference in proximate composition of the muscle; however, an obvious decrease in muscle fiber size and significantly higher hardness, chewiness, and gumminess were observed. Transcriptome results showed that a total of 197 genes were differentially regulated in the dorsal muscle. Down-regulated genes included four genes annotated with myocyte development and 12 transcripts annotated with components of myofibrils. In addition, the FBFG group exhibited significantly lower expression of genes associated with oxygen transport, the mitochondrial respiratory chain, and creatine metabolism, suggesting reduced energy availability in the muscle of the FBFG. Moreover, using western-blotting and enzyme assays, we found decreased protein levels in the mitochondrial electron transport respiratory chain and creatine metabolism activities, as well as increased expression of autophagy marker protein levels, in the muscle of FBFG. Overall, our results suggest that an abnormal energy distribution may exist in grass carps after feeding with faba bean, which is reflected by a mass of fat deposition in the adipose tissue and hepatopancreas and subdued metabolic activity in the muscle.
Collapse
Affiliation(s)
| | | | - Er-meng Yu
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | | | | | | | | | | | | | | | - Jun Xie
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| |
Collapse
|
33
|
Sakushima K, Yoshikawa M, Osaki T, Miyamoto N, Hashimoto T. Moderate hypoxia promotes skeletal muscle cell growth and hypertrophy in C2C12 cells. Biochem Biophys Res Commun 2020; 525:921-927. [PMID: 32173524 DOI: 10.1016/j.bbrc.2020.02.152] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 02/25/2020] [Indexed: 01/25/2023]
Abstract
Although several studies have implied that a hypoxic environment may be a factor that influences muscle hypertrophy, scant attention has been paid to the effect of oxygen molecules on the morphological characteristics of muscle. The purpose of the present study was to examine the effect of semisevere (i.e., 5%) to moderate (i.e., 10% or 15%) hypoxic environments on the morphological characteristics of skeletal muscle and the associated mechanisms. C2C12 skeletal muscle cells were divided into various groups, namely, the normoxia group (20.9% O2) and hypoxia groups (5% O2, 10% O2, and 15% O2), and cell growth and the expression of associated proteins in the hypoxia groups were compared with those in the normoxia group. The myotube diameter and cell differentiation index were determined on day 6 by immunocytochemical analyses. The expression of proteins associated with muscle cell differentiation (MyoD and myogenin) and muscle hypertrophy (mTOR and p70s6K) were analyzed by Western blotting. We found that compared with normoxia, a 5% oxygen environment inhibited differentiation and caused muscle atrophy. However, compared with normoxia, a 10% oxygen environment promoted muscle differentiation, and 10% oxygen and 15% oxygen environments induced muscle hypertrophy. Compared with normoxia, a 10% oxygen environment promoted myogenin and the expression of mTOR, p70s6K, and the metabolic signal AMPK. We concluded that a hypoxic environment, if not too severe, may promote muscle differentiation and hypertrophy by increasing the expression of proteins associated with muscle cell differentiation and hypertrophy.
Collapse
Affiliation(s)
- Koki Sakushima
- Faculty of Sport & Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Maki Yoshikawa
- Faculty of Sport & Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Tomohiko Osaki
- Faculty of Sport & Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan; Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | - Naokazu Miyamoto
- Graduate School of Health and Sports Science, Juntendo University, 1-1 Hiraka-Gakuendai, Inzai, Chiba, 270-1695, Japan
| | - Takeshi Hashimoto
- Faculty of Sport & Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
34
|
Increase in HDAC9 suppresses myoblast differentiation via epigenetic regulation of autophagy in hypoxia. Cell Death Dis 2019; 10:552. [PMID: 31320610 PMCID: PMC6639330 DOI: 10.1038/s41419-019-1763-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/11/2019] [Accepted: 06/19/2019] [Indexed: 02/08/2023]
Abstract
Extremely reduced oxygen (O2) levels are detrimental to myogenic differentiation and multinucleated myotube formation, and chronic exposure to high-altitude hypoxia has been reported to be an important factor in skeletal muscle atrophy. However, how chronic hypoxia causes muscle dysfunction remains unknown. In the present study, we found that severe hypoxia (1% O2) significantly inhibited the function of C2C12 cells (from a myoblast cell line). Importantly, the impairment was continuously manifested even during culture under normoxic conditions for several passages. Mechanistically, we revealed that histone deacetylases 9 (HDAC9), a member of the histone deacetylase family, was significantly increased in C2C12 cells under hypoxic conditions, thereby inhibiting intracellular autophagy levels by directly binding to the promoter regions of Atg7, Beclin1, and LC3. This phenomenon resulted in the sequential dephosphorylation of GSK3β and inactivation of the canonical Wnt pathway, impairing the function of the C2C12 cells. Taken together, our results suggest that hypoxia-induced myoblast dysfunction is due to aberrant epigenetic regulation of autophagy, and our experimental evidence reveals the possible molecular pathogenesis responsible for some muscle diseases caused by chronic hypoxia and suggests a potential therapeutic option.
Collapse
|
35
|
Bensaid S, Fabre C, Fourneau J, Cieniewski-Bernard C. Impact of different methods of induction of cellular hypoxia: focus on protein homeostasis signaling pathways and morphology of C2C12 skeletal muscle cells differentiated into myotubes. J Physiol Biochem 2019; 75:367-377. [PMID: 31267382 DOI: 10.1007/s13105-019-00687-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/15/2019] [Indexed: 12/25/2022]
Abstract
Hypoxia, occurring in several pathologies, has deleterious effects on skeletal muscle, in particular on protein homeostasis. Different induction methods of hypoxia are commonly used in cellular models to investigate the alterations of muscular function consecutive to hypoxic stress. However, a consensus is not clearly established concerning hypoxia induction methodology. Our aim was to compare oxygen deprivation with chemically induced hypoxia using cobalt chloride (CoCl2) or desferrioxamine (DFO) on C2C12 myotubes which were either cultured in hypoxia chamber at an oxygen level of 4% or treated with CoCl2 or DFO. For each method of hypoxia induction, we determined their impact on muscle cell morphology and on expression or activation status of key signaling proteins of synthesis and degradation pathways. The expression of HIF-1α increased whatever the method of hypoxia induction. Myotube diameter and protein content decreased exclusively for C2C12 myotubes submitted to physiological hypoxia (4% O2) or treated with CoCl2. Results were correlated with a hypophosphorylation of key proteins regulated synthesis pathway (Akt, GSK3-β and P70S6K). Similarly, the phosphorylation of FoxO1 decreased and the autophagy-related LC3-II was overexpressed with 4% O2 and CoCl2 conditions. Our results demonstrated that in vitro oxygen deprivation and the use of mimetic agent such as CoCl2, unlike DFO, induced similar responses on myotube morphology and atrophy/hypertrophy markers. Thus, physiological hypoxia or its artificial induction using CoCl2 can be used to understand finely the molecular changes in skeletal muscle cells and to evaluate new therapeutics for hypoxia-related muscle disorders.
Collapse
Affiliation(s)
- Samir Bensaid
- Team Physical Activity, Muscle, Health, University Lille - EA 7369 - URePSSS, 59000, Lille, France.,Research Pole, CHU Lille, 59000, Lille, France
| | - Claudine Fabre
- Team Physical Activity, Muscle, Health, University Lille - EA 7369 - URePSSS, 59000, Lille, France
| | - Julie Fourneau
- Team Physical Activity, Muscle, Health, University Lille - EA 7369 - URePSSS, 59000, Lille, France
| | | |
Collapse
|
36
|
Ross CI, Shute RJ, Ruby BC, Slivka DR. Skeletal Muscle mRNA Response to Hypobaric and Normobaric Hypoxia After Normoxic Endurance Exercise. High Alt Med Biol 2019; 20:141-149. [DOI: 10.1089/ham.2018.0147] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Caleb I. Ross
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| | - Robert J. Shute
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| | - Brent C. Ruby
- Montana Center for Work Physiology and Exercise Metabolism, University of Montana, Missoula, Montana
| | - Dustin R. Slivka
- Exercise Physiology Lab, University of Nebraska at Omaha, Omaha, Nebraska
| |
Collapse
|
37
|
Hori S, Hiramuki Y, Nishimura D, Sato F, Sehara-Fujisawa A. PDH‐mediated metabolic flow is critical for skeletal muscle stem cell differentiation and myotube formation during regeneration in mice. FASEB J 2019; 33:8094-8109. [DOI: 10.1096/fj.201802479r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Shimpei Hori
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| | - Yosuke Hiramuki
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
- Human Biology DivisionFred Hutchinson Cancer Research Center Seattle Washington USA
| | - Daigo Nishimura
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| | - Fuminori Sato
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| | - Atsuko Sehara-Fujisawa
- Department of Growth RegulationInstitute for Frontier Life and Medical SciencesKyoto University Kyoto Japan
| |
Collapse
|
38
|
Yoo YM, Jung EM, Jeung EB. Rapamycin-induced autophagy decreases Myf5 and MyoD proteins in C2C12 myoblast cells. Toxicol In Vitro 2019; 58:132-141. [PMID: 30905858 DOI: 10.1016/j.tiv.2019.03.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022]
Abstract
Rapamycin is an immunosuppressant that inhibits the mammalian or mechanistic target of rapamycin (mTOR) protein kinase and extends lifespan in organisms including mice. Myf5 and MyoD act as muscle-specific transcriptional factors for skeletal muscle differentiation. In this study, we determined whether rapamycin-induced autophagy causes the decrease of Myf5 and MyoD protein in C2C12 myoblast cells. Rapamycin induced a significant increase in the expression of the microtubule-associated protein 1 light chain 3 (LC3) II protein in a dose-dependent manner for 12 h. Rapamycin treatment also significantly increased p-ERK, p-Akt, and catalase expressions, and decreased Mn-SOD expression in a dose-dependent manner. Bax expression was significantly high compared to Bcl-2 expression in a dose-dependent manner of rapamycin for 12 h. For further study of rapamycin-induced autophagy in C2C12 myoblast cells, we investigated rapamycin treatment for 24, 36, and 48 h. Cell viability did not change with rapamycin treatment for 24, 36, and 48 h. Rapamycin-induced LC3-II, Beclin-1, Bax, and Bcl-2 proteins were significantly increased compared to without rapamycin. p-ERK expression increased with rapamycin treatment for 24 and 36 h compared to that without rapamycin, but decreased for 48 h. p-Akt expression decreased with rapamycin treatment for 36 and 48 h compared to that without rapamycin. In the same conditions, rapamycin-induced autophagy significantly reduced the Myf5 and MyoD proteins. Together, these results suggest that rapamycin-induced autophagy results in the decrease of Myf5 and MyoD proteins in C2C12 myoblast cells.
Collapse
Affiliation(s)
- Yeong-Min Yoo
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Eui-Man Jung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
39
|
Baumann M, Gumpold C, Mueller-Felber W, Schoser B, Haberler C, Loescher WN, Rostásy K, Fischer MB, Wanschitz JV. Pattern of myogenesis and vascular repair in early and advanced lesions of juvenile dermatomyositis. Neuromuscul Disord 2018; 28:973-985. [DOI: 10.1016/j.nmd.2018.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/15/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022]
|
40
|
Huang Y, Lai X, Hu L, Lei C, Lan X, Zhang C, Ma Y, Zheng L, Bai Y, Lin F, Chen H. Over‐expression of DEC1 inhibits myogenic differentiation by modulating MyoG activity in bovine satellite cell. J Cell Physiol 2018; 233:9365-9374. [DOI: 10.1002/jcp.26471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/05/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Yongzhen Huang
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| | - Xinsheng Lai
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
- The Laboratory of Synaptic Development and Plasticity, Institute of Life ScienceNanchang UniversityNanchangChina
- School of Life ScienceNanchang UniversityNanchangChina
| | - Linyong Hu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau BiologyChinese Academy of SciencesXiningQinghaiChina
| | - Chuzhao Lei
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| | - Xianyong Lan
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| | - Chunlei Zhang
- Institute of Cellular and Molecular BiologyJiangsu Normal UniversityXuzhouJiangsuChina
| | - Yun Ma
- College of Life Sciences, Xinyang Normal UniversityInstitute for Conservation and Utilization of Agro‐Bioresources in Dabie MountainsXinyangHenanChina
| | - Li Zheng
- Henan University of Animal Husbandry and EconomyZhengzhouHenanChina
| | - Yue‐Yu Bai
- Animal Health Supervision in Henan ProvinceZhengzhouHenanChina
| | - Fengpeng Lin
- Bureau of Animal Husbandry of Biyang CountyBiyangHenanChina
| | - Hong Chen
- Shaanxi Key Laboratory of Molecular Biology for AgricultureCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxiChina
| |
Collapse
|
41
|
Saenz Del Burgo L, Ciriza J, Espona-Noguera A, Illa X, Cabruja E, Orive G, Hernández RM, Villa R, Pedraz JL, Alvarez M. 3D Printed porous polyamide macrocapsule combined with alginate microcapsules for safer cell-based therapies. Sci Rep 2018; 8:8512. [PMID: 29855599 PMCID: PMC5981392 DOI: 10.1038/s41598-018-26869-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/17/2018] [Indexed: 02/06/2023] Open
Abstract
Cell microencapsulation is an attractive strategy for cell-based therapies that allows the implantation of genetically engineered cells and the continuous delivery of de novo produced therapeutic products. However, the establishment of a way to retrieve the implanted encapsulated cells in case the treatment needs to be halted or when cells need to be renewed is still a big challenge. The combination of micro and macroencapsulation approaches could provide the requirements to achieve a proper immunoisolation, while maintaining the cells localized into the body. We present the development and characterization of a porous implantable macrocapsule device for the loading of microencapsulated cells. The device was fabricated in polyamide by selective laser sintering (SLS), with controlled porosity defined by the design and the sintering conditions. Two types of microencapsulated cells were tested in order to evaluate the suitability of this device; erythropoietin (EPO) producing C2C12 myoblasts and Vascular Endothelial Growth Factor (VEGF) producing BHK fibroblasts. Results showed that, even if the metabolic activity of these cells decreased over time, the levels of therapeutic protein that were produced and, importantly, released to the media were stable.
Collapse
Affiliation(s)
- Laura Saenz Del Burgo
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Jesús Ciriza
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Albert Espona-Noguera
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Xavi Illa
- Instituto de Microelectronica de Barcelona (IMB-CNM, CSIC), Campus UAB, 08193 Bellaterra, Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Enric Cabruja
- Instituto de Microelectronica de Barcelona (IMB-CNM, CSIC), Campus UAB, 08193 Bellaterra, Barcelona, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Rosa María Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Rosa Villa
- Instituto de Microelectronica de Barcelona (IMB-CNM, CSIC), Campus UAB, 08193 Bellaterra, Barcelona, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006, Vitoria-Gasteiz, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Mar Alvarez
- Instituto de Microelectronica de Barcelona (IMB-CNM, CSIC), Campus UAB, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
42
|
Rahar B, Chawla S, Pandey S, Bhatt AN, Saxena S. Sphingosine-1-phosphate pretreatment amends hypoxia-induced metabolic dysfunction and impairment of myogenic potential in differentiating C2C12 myoblasts by stimulating viability, calcium homeostasis and energy generation. J Physiol Sci 2018; 68:137-151. [PMID: 28070865 PMCID: PMC10717551 DOI: 10.1007/s12576-016-0518-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/26/2016] [Indexed: 12/19/2022]
Abstract
Sphingosine-1-phosphate (S1P) has a role in transpiration in patho-physiological signaling in skeletal muscles. The present study evaluated the pre-conditioning efficacy of S1P in facilitating differentiation of C2C12 myoblasts under a normoxic/hypoxic cell culture environment. Under normoxia, exogenous S1P significantly promoted C2C12 differentiation as evident from morphometric descriptors and differentiation markers of the mature myotubes, but it could facilitate only partial recovery from hypoxia-induced compromised differentiation. Pretreatment of S1P optimized the myokine secretion, intracellular calcium release and energy generation by boosting the aerobic/anaerobic metabolism and mitochondrial mass. In the hypoxia-exposed cells, there was derangement of the S1PR1-3 expression patterns, while the same could be largely restored with S1P pretreatment. This is being proposed as a plausible underlying mechanism for the observed pro-myogenic efficacy of exogenous S1P preconditioning. The present findings are an invaluable addition to the existing knowledge on the pro-myogenic potential of S1P and may prove beneficial in the field of hypoxia-related myo-pathologies.
Collapse
Affiliation(s)
- Babita Rahar
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Sonam Chawla
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Sanjay Pandey
- Division of Metabolic and Cell Signaling Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Anant Narayan Bhatt
- Division of Metabolic and Cell Signaling Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Shweta Saxena
- Medicinal and Aromatic Plant Division, Defence Institute of High Altitude Research (DIHAR), Defence Research and Development Organization (DRDO), Ministry of Defence, Leh-Ladakh, 194101, Jammu and Kashmir, India.
| |
Collapse
|
43
|
Abstract
Background: p53 is a tumor suppressor protein involved in regulating a wide array of signaling pathways. The role of p53 in the cell is determined by the type of imposed oxidative stress, its intensity and duration. The last decade of research has unravelled a dual nature in the function of p53 in mediating the oxidative stress burden. However, this is dependent on the specific properties of the applied stress and thus requires further analysis. Methods: A systematic review was performed following an electronic search of Pubmed, Google Scholar, and ScienceDirect databases. Articles published in the English language between January 1, 1990 and March 1, 2017 were identified and isolated based on the analysis of p53 in skeletal muscle in both animal and cell culture models. Results: Literature was categorized according to the modality of imposed oxidative stress including exercise, diet modification, exogenous oxidizing agents, tissue manipulation, irradiation, and hypoxia. With low to moderate levels of oxidative stress, p53 is involved in activating pathways that increase time for cell repair, such as cell cycle arrest and autophagy, to enhance cell survival. However, with greater levels of stress intensity and duration, such as with irradiation, hypoxia, and oxidizing agents, the role of p53 switches to facilitate increased cellular stress levels by initiating DNA fragmentation to induce apoptosis, thereby preventing aberrant cell proliferation. Conclusion: Current evidence confirms that p53 acts as a threshold regulator of cellular homeostasis. Therefore, within each modality, the intensity and duration are parameters of the oxidative stressor that must be analyzed to determine the role p53 plays in regulating signaling pathways to maintain cellular health and function in skeletal muscle. Abbreviations: Acadl: acyl-CoA dehydrogenase, long chain; Acadm: acyl-CoA dehydrogenase, C-4 to C-12 straight chain; AIF: apoptosis-inducing factor; Akt: protein kinase B (PKB); AMPK: AMP-activated protein kinase; ATF-4: activating transcription factor 4; ATM: ATM serine/threonine kinase; Bax: BCL2 associated X, apoptosis regulator; Bcl-2: B cell Leukemia/Lymphoma 2 apoptosis regulator; Bhlhe40: basic helix-loop-helix family member e40; BH3: Borane; Bim: bcl-2 interacting mediator of cell death; Bok: Bcl-2 related ovarian killer; COX-IV: cytochrome c oxidase IV; cGMP: Cyclic guanosine monophosphate; c-myc: proto-oncogene protein; Cpt1b: carnitine palmitoyltransferase 1B; Dr5: death receptor 5; eNOS: endothelial nitric oxide synthase; ERK: extracellular regulated MAP kinase; Fas: Fas Cell surface death receptor; FDXR: Ferredoxin Reductase; FOXO3a: forkhead box O3; Gadd45a: growth arrest and DNA damage-inducible 45 alpha; GLS2: glutaminase 2; GLUT 1 and 4: glucose transporter 1(endothelial) and 4 (skeletal muscle); GSH: Glutathione; Hes1: hes family bHLH transcription factor 1; Hey1: hes related family bHLH transcription factor with YRPW motif 1; HIFI-α: hypoxia-inducible factor 1, α-subunit; HK2: Hexokinase 2; HSP70: Heat Shock Protein 70; H2O2: Hydrogen Peroxide; Id2: inhibitor of DNA-binding 2; IGF-1-BP3: Insulin-like growth factor binding protein 3; IL-1β: Interleukin 1 beta; iNOS: inducible nitric oxide synthase; IRS-1: Insulin receptor substrate 1; JNK: c-Jun N-terminal kinases; LY-83583: 6-anilino-5,8-quinolinedione; inhibitor of soluble guanylate cyclase and of cGMP production; Mdm 2/ 4: Mouse double minute 2 homolog (mouse) Mdm4 (humans); mtDNA: mitochondrial DNA; MURF1: Muscle RING-finger protein-1; MyoD: Myogenic differentiation 1; MyoG: myogenin; Nanog: Nanog homeobox; NF-kB: Nuclear factor-κB; NO: nitric oxide; NoxA: phorbol-12-myristate-13-acetate-induced protein 1 (Pmaip1); NRF-1: nuclear respiratory factor 1; Nrf2: Nuclear factor erythroid 2-related factor 2; P21: Cdkn1a cyclin-dependent kinase inhibitor 1A (P21); P38 MAPK: mitogen-activated protein kinases; p53R2: p53 inducible ribonucleotide reductase gene; P66Shc: src homology 2 domain-containing transforming protein C1; PERP: p53 apoptosis effector related to PMP-22; PGC-1α: Peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PGM: phosphoglucomutase; PI3K: Phosphatidylinositol-4,5-bisphosphate 3-kinase; PKCβ: protein kinase c beta; PTEN: phosphatase and tensin homolog; PTIO: 2-phenyl-4, 4, 5, 5,-tetramethylimidazoline-1-oxyl 3-oxide (PTIO) has been used as a nitric oxide (NO) scavenger; Puma: The p53 upregulated modulator of apoptosis; PW1: paternally expressed 3 (Peg3); RNS: Reactive nitrogen species; SIRT1: sirtuin 1; SCO2: cytochrome c oxidase assembly protein; SOD2: superoxide dismutase 2; Tfam: transcription factor A mitochondrial; TIGAR: Trp53 induced glycolysis repulatory phosphatase; TNF-a: tumor necrosis factor a; TRAF2: TNF receptor associated factor 2; TRAIL: type II transmembrane protein.
Collapse
Affiliation(s)
- Kaitlyn Beyfuss
- a School of Kinesiology and Health Sciences , York University , Toronto , Canada
| | - David A Hood
- a School of Kinesiology and Health Sciences , York University , Toronto , Canada
| |
Collapse
|
44
|
Selvaraj S, Oh JH, Spanel R, Länger F, Han HY, Lee EH, Yoon S, Borlak J. The pathogenesis of diclofenac induced immunoallergic hepatitis in a canine model of liver injury. Oncotarget 2017; 8:107763-107824. [PMID: 29296203 PMCID: PMC5746105 DOI: 10.18632/oncotarget.21201] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/31/2017] [Indexed: 12/19/2022] Open
Abstract
Hypersensitivity to non-steroidal anti-inflammatory drugs is a common adverse drug reaction and may result in serious inflammatory reactions of the liver. To investigate mechanism of immunoallergic hepatitis beagle dogs were given 1 or 3 mg/kg/day (HD) oral diclofenac for 28 days. HD diclofenac treatment caused liver function test abnormalities, reduced haematocrit and haemoglobin but induced reticulocyte, WBC, platelet, neutrophil and eosinophil counts. Histopathology evidenced hepatic steatosis and glycogen depletion, apoptosis, acute lobular hepatitis, granulomas and mastocytosis. Whole genome scans revealed 663 significantly regulated genes of which 82, 47 and 25 code for stress, immune response and inflammation. Immunopathology confirmed strong induction of IgM, the complement factors C3&B, SAA, SERPING1 and others of the classical and alternate pathway. Alike, marked expression of CD205 and CD74 in Kupffer cells and lymphocytes facilitate antigen presentation and B-cell differentiation. The highly induced HIF1A and KLF6 protein expression in mast cells and macrophages sustain inflammation. Furthermore, immunogenomics discovered 24, 17, 6 and 11 significantly regulated marker genes to hallmark M1/M2 polarized macrophages, lymphocytic and granulocytic infiltrates; note, the latter was confirmed by CAE staining. Other highly regulated genes included alpha-2-macroglobulin, CRP, hepcidin, IL1R1, S100A8 and CCL20. Diclofenac treatment caused unprecedented induction of myeloperoxidase in macrophages and oxidative stress as shown by SOD1/SOD2 immunohistochemistry. Lastly, bioinformatics defined molecular circuits of inflammation and consisted of 161 regulated genes. Altogether, the mechanism of diclofenac induced liver hypersensitivity reactions involved oxidative stress, macrophage polarization, mastocytosis, complement activation and an erroneous programming of the innate and adaptive immune system.
Collapse
Affiliation(s)
- Saravanakumar Selvaraj
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany.,Institute of Pathology, 41747 Viersen, Germany
| | - Florian Länger
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Hyoung-Yun Han
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Eun-Hee Lee
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Seokjoo Yoon
- Department of Predictive Toxicology, Korea Institute of Toxicology, 34114 Gajeong-ro, Yuseong, Daejeon, Republic of Korea
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
45
|
IGF-1 Attenuates Hypoxia-Induced Atrophy but Inhibits Myoglobin Expression in C2C12 Skeletal Muscle Myotubes. Int J Mol Sci 2017; 18:ijms18091889. [PMID: 28862673 PMCID: PMC5618538 DOI: 10.3390/ijms18091889] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/23/2017] [Accepted: 08/29/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic hypoxia is associated with muscle wasting and decreased oxidative capacity. By contrast, training under hypoxia may enhance hypertrophy and increase oxidative capacity as well as oxygen transport to the mitochondria, by increasing myoglobin (Mb) expression. The latter may be a feasible strategy to prevent atrophy under hypoxia and enhance an eventual hypertrophic response to anabolic stimulation. Mb expression may be further enhanced by lipid supplementation. We investigated individual and combined effects of hypoxia, insulin-like growth factor (IGF)-1 and lipids, in mouse skeletal muscle C2C12 myotubes. Differentiated C2C12 myotubes were cultured for 24 h under 20%, 5% and 2% oxygen with or without IGF-1 and/or lipid treatment. In culture under 20% oxygen, IGF-1 induced 51% hypertrophy. Hypertrophy was only 32% under 5% and abrogated under 2% oxygen. This was not explained by changes in expression of genes involved in contractile protein synthesis or degradation, suggesting a reduced rate of translation rather than of transcription. Myoglobin mRNA expression increased by 75% under 5% O2 but decreased by 50% upon IGF-1 treatment under 20% O2, compared to control. Inhibition of mammalian target of rapamycin (mTOR) activation using rapamycin restored Mb mRNA expression to control levels. Lipid supplementation had no effect on Mb gene expression. Thus, IGF-1-induced anabolic signaling can be a strategy to improve muscle size under mild hypoxia, but lowers Mb gene expression.
Collapse
|
46
|
Redondo PA, Pavlou M, Loizidou M, Cheema U. Elements of the niche for adult stem cell expansion. J Tissue Eng 2017; 8:2041731417725464. [PMID: 28890779 PMCID: PMC5574483 DOI: 10.1177/2041731417725464] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 07/18/2017] [Indexed: 12/21/2022] Open
Abstract
Adult stem cells are crucial for tissue homeostasis. These cells reside within exclusive locations in tissues, termed niches, which protect adult stem cell fidelity and regulate their many functions through biophysical-, biochemical- and cellular-mediated mechanisms. There is a growing understanding of how these mechanisms and their components contribute towards maintaining stem cell quiescence, self-renewal, expansion and differentiation patterns. In vitro expansion of adult stem cells is a powerful tool for understanding stem cell biology, and for tissue engineering and regenerative medicine applications. However, it is technically challenging, since adult stem cell removal from their native microenvironment has negative repercussions on their sustainability. In this review, we overview specific elements of the biomimetic niche and how recreating such elements can help in vitro propagation of adult stem cells.
Collapse
Affiliation(s)
- Patricia A Redondo
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Marina Pavlou
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| | - Marilena Loizidou
- Division of Surgery and Interventional Science, University College London, London, UK
| | - Umber Cheema
- Institute of Orthopaedics & Musculoskeletal Science, University College London, London, UK
| |
Collapse
|
47
|
Ballester-Beltrán J, Trujillo S, Alakpa EV, Compañ V, Gavara R, Meek D, West CC, Péault B, Dalby MJ, Salmerón-Sánchez M. Confined Sandwichlike Microenvironments Tune Myogenic Differentiation. ACS Biomater Sci Eng 2017; 3:1710-1718. [PMID: 28824958 PMCID: PMC5558191 DOI: 10.1021/acsbiomaterials.7b00109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/09/2017] [Indexed: 12/29/2022]
Abstract
Sandwichlike (SW) cultures are engineered as a multilayer technology to simultaneously stimulate dorsal and ventral cell receptors, seeking to mimic cell adhesion in three-dimensional (3D) environments in a reductionist manner. The effect of this environment on cell differentiation was investigated for several cell types cultured in standard growth media, which promotes proliferation on two-dimensional (2D) surfaces and avoids any preferential differentiation. First, murine C2C12 myoblasts showed specific myogenic differentiation. Human mesenchymal stem cells (hMSCs) of adipose and bone marrow origin, which can differentiate toward a wider variety of lineages, showed again myodifferentiation. Overall, this study shows myogenic differentiation in normal growth media for several cell types under SW conditions, avoiding the use of growth factors and cytokines, i.e., solely by culturing cells within the SW environment. Mechanistically, it provides further insights into the balance between integrin adhesion to the dorsal substrate and the confinement imposed by the SW system.
Collapse
Affiliation(s)
- José Ballester-Beltrán
- Division
of Biomedical Engineering, School of Engineering, University of Glasgow. Rankine Building, Oakfield Avenue, Glasgow G12 8LT, United Kingdom
| | - Sara Trujillo
- Division
of Biomedical Engineering, School of Engineering, University of Glasgow. Rankine Building, Oakfield Avenue, Glasgow G12 8LT, United Kingdom
| | - Enateri V. Alakpa
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology, University of Glasgow. Joseph Black Building, University Avenue, Glasgow G12 8QQ, United Kingdom
| | - Vicente Compañ
- Escuela
Técnica Superior de Ingenieros Industriales, Departamento de
Termodinámica Aplicada, Universitat
Politècnica de València, Camino de Vera s/n, Valencia, Valencia 46022, Spain
| | - Rafael Gavara
- Instituto
de Agroquímica y Tecnología de Alimentos. Consejo Superior
de Investigaciones Científicas (IATA-CSIC), Departamento de Investigación: Conservación y Calidad
de Alimentos,Calle Agustín
Escardino 7, Paterna, Valencia 46980, Spain
| | - Dominic Meek
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology, University of Glasgow. Joseph Black Building, University Avenue, Glasgow G12 8QQ, United Kingdom
| | - Christopher C. West
- Centre for
Regenerative Medicine and Centre for Cardiovascular Science, University of Edinburgh. 47 Little France Crescent, Edinburgh EH16 4TJ, United
Kingdom
| | - Bruno Péault
- Centre for
Regenerative Medicine and Centre for Cardiovascular Science, University of Edinburgh. 47 Little France Crescent, Edinburgh EH16 4TJ, United
Kingdom
| | - Matthew J. Dalby
- Centre
for Cell Engineering, Institute of Molecular, Cell and Systems Biology, University of Glasgow. Joseph Black Building, University Avenue, Glasgow G12 8QQ, United Kingdom
| | - Manuel Salmerón-Sánchez
- Division
of Biomedical Engineering, School of Engineering, University of Glasgow. Rankine Building, Oakfield Avenue, Glasgow G12 8LT, United Kingdom
| |
Collapse
|
48
|
Abstract
One of the differences between normal and cancer cells is lower pH of the extracellular space in tumors. Low pH in the extracellular space activates proteases and stimulates tumor invasion and metastasis. Tumor cells display higher level of the HIF1α transcription factor that promotes cell switch from mitochondrial respiration to glycolysis. The terminal product of glycolysis is lactate. Lactate formation from pyruvate is catalyzed by the specific HIF1α-dependent isoform of lactate dehydrogenase A. Because lactate accumulation is deleterious for the cell, it is actively exported by monocarboxylate transporters. Lactate is cotransported with proton, which acidifies the extracellular space. Another protein that contributes to proton concentration increase in the extracellular space is tumor-specific HIF1α-dependent carbonic anhydrase IX, which generates a proton in the reaction between carbon dioxide and water. The activity of Na+/H+ exchanger (another protein pump) is stimulated by stress factors (e.g. osmotic shock) and proliferation stimuli. This review describes the mechanisms of proton pump activation and reviews results of studies on effects of various proton pump inhibitors on tumor functioning and growth in cell culture and in vivo. The prospects of combined application of proton pump inhibitors and cytostatics in cancer therapy are discussed.
Collapse
Affiliation(s)
- V A Kobliakov
- Blokhin Russian Cancer Research Center, Russian Ministry of Health, Moscow, 115478, Russia.
| |
Collapse
|
49
|
Martin NRW, Aguilar-Agon K, Robinson GP, Player DJ, Turner MC, Myers SD, Lewis MP. Hypoxia Impairs Muscle Function and Reduces Myotube Size in Tissue Engineered Skeletal Muscle. J Cell Biochem 2017; 118:2599-2605. [PMID: 28294416 PMCID: PMC5518201 DOI: 10.1002/jcb.25982] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 02/21/2017] [Indexed: 12/21/2022]
Abstract
Contemporary tissue engineered skeletal muscle models display a high degree of physiological accuracy compared with native tissue, and therefore may be excellent platforms to understand how various pathologies affect skeletal muscle. Chronic obstructive pulmonary disease (COPD) is a lung disease which causes tissue hypoxia and is characterized by muscle fiber atrophy and impaired muscle function. In the present study we exposed engineered skeletal muscle to varying levels of oxygen (O2; 21–1%) for 24 h in order to see if a COPD like muscle phenotype could be recreated in vitro, and if so, at what degree of hypoxia this occurred. Maximal contractile force was attenuated in hypoxia compared to 21% O2; with culture at 5% and 1% O2 causing the most pronounced effects with 62% and 56% decrements in force, respectively. Furthermore at these levels of O2, myotubes within the engineered muscles displayed significant atrophy which was not seen at higher O2 levels. At the molecular level we observed increases in mRNA expression of MuRF‐1 only at 1% O2 whereas MAFbx expression was elevated at 10%, 5%, and 1% O2. In addition, p70S6 kinase phosphorylation (a downstream effector of mTORC1) was reduced when engineered muscle was cultured at 1% O2, with no significant changes seen above this O2 level. Overall, these data suggest that engineered muscle exposed to O2 levels of ≤5% adapts in a manner similar to that seen in COPD patients, and thus may provide a novel model for further understanding muscle wasting associated with tissue hypoxia. J. Cell. Biochem. 118: 2599–2605, 2017. © 2017 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neil R W Martin
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Kathyrn Aguilar-Agon
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - George P Robinson
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Darren J Player
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Mark C Turner
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Stephen D Myers
- Department of Sport and Exercise Sciences, University of Chichester, Chichester, UK
| | - Mark P Lewis
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| |
Collapse
|
50
|
Cirillo F, Resmini G, Ghiroldi A, Piccoli M, Bergante S, Tettamanti G, Anastasia L. Activation of the hypoxia‐inducible factor 1a promotes myogenesis through the noncanonical Wnt pathway, leading to hypertrophic myotubes. FASEB J 2017; 31:2146-2156. [DOI: 10.1096/fj.201600878r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/23/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Federica Cirillo
- Laboratory of Stem Cells for Tissue EngineeringIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San DonatoMilanItaly
| | - Giulia Resmini
- Laboratory of Stem Cells for Tissue EngineeringIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San DonatoMilanItaly
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue EngineeringIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San DonatoMilanItaly
| | - Marco Piccoli
- Laboratory of Stem Cells for Tissue EngineeringIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San DonatoMilanItaly
| | - Sonia Bergante
- Laboratory of Stem Cells for Tissue EngineeringIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San DonatoMilanItaly
| | - Guido Tettamanti
- Laboratory of Stem Cells for Tissue EngineeringIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San DonatoMilanItaly
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue EngineeringIstituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San DonatoMilanItaly
- Department of Biomedical Sciences for HealthUniversity of MilanMilanItaly
| |
Collapse
|