1
|
Förster PM, Hogenkamp J, Pottgießer MF, Binsch C, Humpert AD, Brügge CL, Deatc MI, Ensenauer R, Chadt A, Thoresen GH, Ouwens DM, Hartwig S, Lehr S, Al-Hasani H. High-resolution analyses of the secretomes from murine C2C12 cells and primary human skeletal muscle cells reveal distinct differences in contraction-regulated myokine secretion. Front Physiol 2025; 16:1549316. [PMID: 40200984 PMCID: PMC11975866 DOI: 10.3389/fphys.2025.1549316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/24/2025] [Indexed: 04/10/2025] Open
Abstract
Myokines released by skeletal muscle in response to contraction may contribute to the health-promoting effects of exercise. Previous studies with cultured rodent and human myotubes have revealed highly complex patterns of myokine secretion. However, the commonalities and differences in the secretory response of the different cell models have not been explored, limiting the interpretation of these results. In the present study, we performed a comprehensive analysis of contraction-regulated secretomes using the most commonly used skeletal muscle cell models, cultured murine C2C12 myotubes and satellite cell-derived primary human myotubes (HSkMC). The cells were subjected to low-frequency electrical pulse stimulation (EPS) for 6 h followed by high-resolution mass spectrometry analysis of secreted proteins in the culture medium. We identified 5,710 and 3,285 proteins in the secretomes of C2C12 myotubes and HSkMC, with 80% of human myokines also detected in the murine secretome. Additionally, we found 518 and 336 secreted proteins that were differentially regulated during contraction in murine and human cells, respectively, along with 1,440 and 385 previously unknown potential myokines secreted by murine and human myotubes. Bioinformatic prediction analyses revealed that the majority of the newly identified myokines were secreted via unconventional protein secretion pathways (UPS) in the murine secretome, whereas most novel proteins in the human secretome were secreted via the classical endoplasmic reticulum (ER)-to-Golgi pathway. Moreover, ontology analysis indicates cell type-specific differences in cellular compartments involved in myokine secretion. Collectively, our results provide a comprehensive overview of the secretomes of two of the most commonly used cell models and may provide guidance for further studies of myokines.
Collapse
Affiliation(s)
- Pia Marlene Förster
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Julian Hogenkamp
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Moira Fee Pottgießer
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Christian Binsch
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Awovi Didi Humpert
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Carolin Laura Brügge
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Michelle Isabel Deatc
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Regina Ensenauer
- Institute of Child Nutrition, Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Karlsruhe, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - G. Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - D. Margriet Ouwens
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Sonja Hartwig
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Stefan Lehr
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| |
Collapse
|
2
|
Liu X, Yao Z, Zhang L, Shyh‐Chang N. Muscle-Derived Bioactive Factors: MyoEVs and Myokines. Cell Prolif 2025; 58:e13801. [PMID: 39737773 PMCID: PMC11882754 DOI: 10.1111/cpr.13801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/01/2025] Open
Abstract
Overview of the functions and applications of myokines and MyoEVs.
Collapse
Affiliation(s)
- Xupeng Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Ziyue Yao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Liping Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| | - Ng Shyh‐Chang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of Zoology, Chinese Academy of SciencesBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
| |
Collapse
|
3
|
Bu A, Afghah F, Castro N, Bawa M, Kohli S, Shah K, Rios B, Butty V, Raman R. Actuating Extracellular Matrices Decouple the Mechanical and Biochemical Effects of Muscle Contraction on Motor Neurons. Adv Healthc Mater 2025; 14:e2403712. [PMID: 39523700 PMCID: PMC11874633 DOI: 10.1002/adhm.202403712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Emerging in vivo evidence suggests that repeated muscle contraction, or exercise, impacts peripheral nerves. However, the difficulty of isolating the muscle-specific impact on motor neurons in vivo, as well as the inability to decouple the biochemical and mechanical impacts of muscle contraction in this setting, motivates investigating this phenomenon in vitro. This study demonstrates that tuning the mechanical properties of fibrin enables longitudinal culture of highly contractile skeletal muscle monolayers, enabling functional characterization of and long-term secretome harvesting from exercised tissues. Motor neurons stimulated with exercised muscle-secreted factors significantly upregulate neurite outgrowth and migration, with an effect size dependent on muscle contraction intensity. Actuating magnetic microparticles embedded within fibrin hydrogels enable dynamically stretching motor neurons and non-invasively mimicking the mechanical effects of muscle contraction. Interestingly, axonogenesis is similarly upregulated in both mechanically and biochemically stimulated motor neurons, but RNA sequencing reveals different transcriptomic signatures between groups, with biochemical stimulation having a greater impact on cell signaling related to axonogenesis and synapse maturation. This study leverages actuating extracellular matrices to robustly validate a previously hypothesized role for muscle contraction in regulating motor neuron growth and maturation from the bottom-up through both mechanical and biochemical signaling.
Collapse
Affiliation(s)
- Angel Bu
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ferdows Afghah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Nicolas Castro
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Maheera Bawa
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sonika Kohli
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Karina Shah
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Brandon Rios
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Vincent Butty
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ritu Raman
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
4
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
5
|
Xie Y, Liu X, Liu W, Carr LR, Lee LP, Imai N, Ortlund EA, Cohen DE. Activity and phosphatidylcholine transfer protein interactions of skeletal muscle thioesterase Them2 enable hepatic steatosis and insulin resistance. J Biol Chem 2024; 300:107855. [PMID: 39369989 PMCID: PMC11570472 DOI: 10.1016/j.jbc.2024.107855] [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: 08/04/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024] Open
Abstract
Thioesterase superfamily member 2 (Them2), a long-chain fatty acyl-CoA thioesterase that is highly expressed in oxidative tissues, interacts with phosphatidylcholine transfer protein (PC-TP) to regulate hepatic lipid and glucose metabolism and to suppress insulin signaling. High-fat diet-fed mice lacking Them2 globally or specifically in skeletal muscle, but not liver, exhibit reduced hepatic steatosis and insulin resistance. Here, we report that the capacity of Them2 in skeletal muscle to promote hepatic steatosis and insulin resistance depends on both its catalytic activity and interaction with PC-TP. Two residues of Them2 catalytic site were mutated (N50A/D65A) to produce the inactive enzyme while maintaining its homotetrameric structure and interaction with PC-TP. Restoration of skeletal muscle expression in Them2-/- mice using recombinant adeno-associated virus revealed that WT, but not N50A/D65A Them2, promoted high-fat diet-induced weight gain and hepatic steatosis. This was accompanied by greater impairment of insulin sensitivity in WT than N50A/D65A Them2. Pharmacological inhibition or genetic ablation of PC-TP attenuated these effects. In reductionist experiments, conditioned medium collected from WT primary cultured myotubes promoted excess lipid accumulation in oleic acid-treated primary cultured hepatocytes relative to Them2-/- myotubes, which was attributable to secreted extracellular vesicles. Reconstitution of Them2 expression in Them2-/- myotubes affirmed the requirements for catalytic activity and PC-TP interactions for extracellular vesicles to promote lipid accumulation in hepatocytes. These studies provide valuable mechanistic insights, whereby Them2 in skeletal muscle promotes hepatic steatosis and establish both Them2 and PC-TP as attractive targets for managing metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Yang Xie
- Division of Gastroenterology, Hepatology & Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xu Liu
- Department of Biochemistry, Emory University, Atlanta, Georgia, USA
| | - Wenpeng Liu
- Division of Renal Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Logan R Carr
- Division of Gastroenterology, Hepatology & Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Luke P Lee
- Division of Renal Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Norihiro Imai
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Eric A Ortlund
- Department of Biochemistry, Emory University, Atlanta, Georgia, USA
| | - David E Cohen
- Division of Gastroenterology, Hepatology & Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
6
|
Vantaggiato L, Landi C, Shaba E, Rossi D, Sorrentino V, Bini L. Protein Extraction Methods Suitable for Muscle Tissue Proteomic Analysis. Proteomes 2024; 12:27. [PMID: 39449499 PMCID: PMC11503273 DOI: 10.3390/proteomes12040027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Muscle tissue is one of the most dynamic and plastic tissues of the mammalian body and covers different roles, such as force generation and metabolic control. Muscular proteomics provides an important opportunity to reveal the molecular mechanisms behind muscle pathophysiology. To ensure successful proteomic analysis, it is necessary to have an efficient and reproducible protein extraction method. This study aimed to evaluate the efficacy of two different extraction protocols of muscle samples for two-dimensional gel electrophoresis. In particular, mouse muscle proteins were extracted by an SDS-based buffer (Method A) and by a UREA/CHAPS/DTE/TRIS solution (Method B). The efficacies of the methods were assessed by performing an image analysis of the 2DE gels and by statistical and multivariate analyses. The 2DE gels in both preparations showed good resolution and good spot overlapping. Methods A and B produced 2DE gels with different means of total spots, higher for B. Image analysis showed different patterns of protein abundance between the protocols. The results showed that the two methods extract and solubilize proteins with different chemical-physical characteristics and different cellular localizations. These results attest the efficacy and reproducibility of both protein extraction methods, which can be parallelly applied for comprehensive proteomic profiling of muscle tissue.
Collapse
Affiliation(s)
- Lorenza Vantaggiato
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| | - Claudia Landi
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| | - Enxhi Shaba
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (D.R.); (V.S.)
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (D.R.); (V.S.)
| | - Luca Bini
- Functional Proteomics Lab., Department Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (L.V.); (E.S.); (L.B.)
| |
Collapse
|
7
|
Boss-Kennedy A, Kim D, Barai P, Maldonado C, Reyes-Ordoñez A, Chen J. Muscle cell-derived Ccl8 is a negative regulator of skeletal muscle regeneration. FASEB J 2024; 38:e23841. [PMID: 39051762 PMCID: PMC11279459 DOI: 10.1096/fj.202400184r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024]
Abstract
Skeletal muscles undergo robust regeneration upon injury, and infiltrating immune cells play a major role in not only clearing damaged tissues but also regulating the myogenic process through secreted cytokines. Chemokine C-C motif ligand 8 (Ccl8), along with Ccl2 and Ccl7, has been reported to mediate inflammatory responses to suppress muscle regeneration. Ccl8 is also expressed by muscle cells, but a role of the muscle cell-derived Ccl8 in myogenesis has not been reported. In this study, we found that knockdown of Ccl8, but not Ccl2 or Ccl7, led to increased differentiation of C2C12 myoblasts. Analysis of existing single-cell transcriptomic datasets revealed that both immune cells and muscle stem cells (MuSCs) in regenerating muscles express Ccl8, with the expression by MuSCs at a much lower level, and that the temporal patterns of Ccl8 expression were different in MuSCs and macrophages. To probe a function of muscle cell-derived Ccl8 in vivo, we utilized a mouse system in which Cas9 was expressed in Pax7+ myogenic progenitor cells (MPCs) and Ccl8 gene editing was induced by AAV9-delivered sgRNA. Depletion of Ccl8 in Pax7+ MPCs resulted in accelerated muscle regeneration after barium chloride-induced injury in both young and middle-aged mice, and intramuscular administration of a recombinant Ccl8 reversed the phenotype. Accelerated regeneration was also observed when Ccl8 was depleted in Myf5+ or MyoD+ MPCs by similar approaches. Our results suggest that muscle cell-derived Ccl8 plays a unique role in regulating the initiation of myogenic differentiation during injury-induced muscle regeneration.
Collapse
Affiliation(s)
- A Boss-Kennedy
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - D Kim
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - P Barai
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - C Maldonado
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - A Reyes-Ordoñez
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - J Chen
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
8
|
Kump DS. Mechanisms Underlying the Rarity of Skeletal Muscle Cancers. Int J Mol Sci 2024; 25:6480. [PMID: 38928185 PMCID: PMC11204341 DOI: 10.3390/ijms25126480] [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: 05/07/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Skeletal muscle (SKM), despite comprising ~40% of body mass, rarely manifests cancer. This review explores the mechanisms that help to explain this rarity, including unique SKM architecture and function, which prohibits the development of new cancer as well as negates potential metastasis to SKM. SKM also presents a unique immune environment that may magnify the anti-tumorigenic effect. Moreover, the SKM microenvironment manifests characteristics such as decreased extracellular matrix stiffness and altered lactic acid, pH, and oxygen levels that may interfere with tumor development. SKM also secretes anti-tumorigenic myokines and other molecules. Collectively, these mechanisms help account for the rarity of SKM cancer.
Collapse
Affiliation(s)
- David S Kump
- Department of Biological Sciences, Winston-Salem State University, 601 Martin Luther King Jr. Dr., Winston-Salem, NC 27110, USA
| |
Collapse
|
9
|
Chen ZT, Weng ZX, Lin JD, Meng ZX. Myokines: metabolic regulation in obesity and type 2 diabetes. LIFE METABOLISM 2024; 3:loae006. [PMID: 39872377 PMCID: PMC11749576 DOI: 10.1093/lifemeta/loae006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/23/2024] [Accepted: 02/29/2024] [Indexed: 01/30/2025]
Abstract
Skeletal muscle plays a vital role in the regulation of systemic metabolism, partly through its secretion of endocrine factors which are collectively known as myokines. Altered myokine levels are associated with metabolic diseases, such as type 2 diabetes (T2D). The significance of interorgan crosstalk, particularly through myokines, has emerged as a fundamental aspect of nutrient and energy homeostasis. However, a comprehensive understanding of myokine biology in the setting of obesity and T2D remains a major challenge. In this review, we discuss the regulation and biological functions of key myokines that have been extensively studied during the past two decades, namely interleukin 6 (IL-6), irisin, myostatin (MSTN), growth differentiation factor 11 (GDF11), fibroblast growth factor 21 (FGF21), apelin, brain-derived neurotrophic factor (BDNF), meteorin-like (Metrnl), secreted protein acidic and rich in cysteine (SPARC), β-aminoisobutyric acid (BAIBA), Musclin, and Dickkopf 3 (Dkk3). Related to these, we detail the role of exercise in myokine expression and secretion together with their contributions to metabolic physiology and disease. Despite significant advancements in myokine research, many myokines remain challenging to measure accurately and investigate thoroughly. Hence, new research techniques and detection methods should be developed and rigorously tested. Therefore, developing a comprehensive perspective on myokine biology is crucial, as this will likely offer new insights into the pathophysiological mechanisms underlying obesity and T2D and may reveal novel targets for therapeutic interventions.
Collapse
Affiliation(s)
- Zhi-Tian Chen
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Zhejiang University-University of Edinburgh Institute (ZJE), School of Medicine, Zhejiang University, Haining, Zhejiang 314400, China
| | - Zhi-Xuan Weng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Zhuo-Xian Meng
- Department of Pathology and Pathophysiology and Department of Cardiology of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
10
|
Chen X, Song X, Li J, Wang J, Yan Y, Yang F. Integrated proteomic, phosphoproteomic, and N-glycoproteomic analyses of small extracellular vesicles from C2C12 myoblasts identify specific PTM patterns in ligand-receptor interactions. Cell Commun Signal 2024; 22:273. [PMID: 38755675 PMCID: PMC11097525 DOI: 10.1186/s12964-024-01640-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/28/2024] [Indexed: 05/18/2024] Open
Abstract
Small extracellular vesicles (sEVs) are important mediators of intercellular communication by transferring of functional components (proteins, RNAs, and lipids) to recipient cells. Some PTMs, including phosphorylation and N-glycosylation, have been reported to play important role in EV biology, such as biogenesis, protein sorting and uptake of sEVs. MS-based proteomic technology has been applied to identify proteins and PTM modifications in sEVs. Previous proteomic studies of sEVs from C2C12 myoblasts, an important skeletal muscle cell line, focused on identification of proteins, but no PTM information on sEVs proteins is available.In this study, we systematically analyzed the proteome, phosphoproteome, and N-glycoproteome of sEVs from C2C12 myoblasts with LC-MS/MS. In-depth analyses of the three proteomic datasets revealed that the three proteomes identified different catalogues of proteins, and PTMomic analysis could expand the identification of cargos in sEVs. At the proteomic level, a high percentage of membrane proteins, especially tetraspanins, was identified. The sEVs-derived phosphoproteome had a remarkably high level of tyrosine-phosphorylated sites. The tyrosine-phosphorylated proteins might be involved with EPH-Ephrin signaling pathway. At the level of N-glycoproteomics, several glycoforms, such as complex N-linked glycans and sialic acids on glycans, were enriched in sEVs. Retrieving of the ligand-receptor interaction in sEVs revealed that extracellular matrix (ECM) and cell adhesion molecule (CAM) represented the most abundant ligand-receptor pairs in sEVs. Mapping the PTM information on the ligands and receptors revealed that N-glycosylation mainly occurred on ECM and CAM proteins, while phosphorylation occurred on different categories of receptors and ligands. A comprehensive PTM map of ECM-receptor interaction and their components is also provided.In summary, we conducted a comprehensive proteomic and PTMomic analysis of sEVs of C2C12 myoblasts. Integrated proteomic, phosphoproteomic, and N-glycoproteomic analysis of sEVs might provide some insights about their specific uptake mechanism.
Collapse
Affiliation(s)
- Xiulan Chen
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xi Song
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaran Li
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jifeng Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yumeng Yan
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Fuquan Yang
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
11
|
Yu W, Yao Y, Ye N, Zhao Y, Ye Z, Wei W, Zhang L, Chen J. The myokine CCL5 recruits subcutaneous preadipocytes and promotes intramuscular fat deposition in obese mice. Am J Physiol Cell Physiol 2024; 326:C1320-C1333. [PMID: 38497114 DOI: 10.1152/ajpcell.00591.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Intramuscular fat (IMF) refers to the lipid stored in skeletal muscle tissue. The number and size of intramuscular adipocytes are the primary factors that regulate IMF content. Intramuscular adipocytes can be derived from either in situ or ectopic migration. In this study, it was discovered that the regulation of IMF levels is achieved through the chemokine (C-C motif) ligand 5 (CCL5)/chemokine (C-C motif) receptor 5 (CCR5) pathway by modulating adipocyte migration. In coculture experiments, C2C12 myotubes were more effective in promoting the migration of 3T3-L1 preadipocytes than C2C12 myoblasts, along with increasing CCL5. Correspondingly, overexpressing the CCR5, one of the receptors of CCL5, in 3T3-L1 preadipocytes facilitated their migration. Conversely, the application of the CCL5/CCR5 inhibitor, MARAVIROC (MVC), reduced this migration. In vivo, transplanted experiments of subcutaneous adipose tissue (SCAT) from transgenic mice expressing green fluorescent protein (GFP) provided evidence that injecting recombinant CCL5 (rCCL5) into skeletal muscle promotes the migration of subcutaneous adipocytes to the skeletal muscle. The level of CCL5 in skeletal muscle increased with obesity. Blocking the CCL5/CCR5 axis by MVC inhibited IMF deposition, whereas elevated skeletal muscle CCL5 promoted IMF deposition in obese mice. These results establish a link between the IMF and the CCL5/CCR5 pathway, which could have a potential application for modulating IMF through adipocyte migration.NEW & NOTEWORTHY C2C12 myotubes attract 3T3-L1 preadipocyte migration regulated by the chemokine (C-C motif) ligand 5 (CCL5)/ chemokine (C-C motif) receptor 5 (CCR5) axis. High levels of skeletal muscle-specific CCL5 promote the migration of subcutaneous adipocytes to skeletal muscle and induce the intramuscular fat (IMF) content.
Collapse
Affiliation(s)
- Wensai Yu
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Yao Yao
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Nanwei Ye
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Yuelei Zhao
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Zijian Ye
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Wei Wei
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Lifan Zhang
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Jie Chen
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| |
Collapse
|
12
|
Fard D, Barbiera A, Dobrowolny G, Tamagnone L, Scicchitano BM. Semaphorins: Missing Signals in Age-dependent Alteration of Neuromuscular Junctions and Skeletal Muscle Regeneration. Aging Dis 2024; 15:517-534. [PMID: 37728580 PMCID: PMC10917540 DOI: 10.14336/ad.2023.0801] [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: 05/03/2023] [Accepted: 08/01/2023] [Indexed: 09/21/2023] Open
Abstract
Skeletal muscle is characterized by a remarkable capacity to rearrange after physiological changes and efficiently regenerate. However, during aging, extensive injury, or pathological conditions, the complete regenerative program is severely affected, with a progressive loss of muscle mass and function, a condition known as sarcopenia. The compromised tissue repair program is attributable to the gradual depletion of stem cells and to altered regulatory signals. Defective muscle regeneration can severely affect re-innervation by motor axons, and neuromuscular junctions (NMJs) development, ultimately leading to skeletal muscle atrophy. Defects in NMJ formation and maintenance occur physiologically during aging and are responsible for the pathogenesis of several neuromuscular disorders. However, it is still largely unknown how neuromuscular connections are restored on regenerating fibers. It has been suggested that attractive and repelling signals used for axon guidance could be implicated in this process; in particular, guidance molecules called semaphorins play a key role. Semaphorins are a wide family of extracellular regulatory signals with a multifaceted role in cell-cell communication. Originally discovered as axon guidance factors, they have been implicated in cancer progression, embryonal organogenesis, skeletal muscle innervation, and other physiological and developmental functions in different tissues. In particular, in skeletal muscle, specific semaphorin molecules are involved in the restoration and remodeling of the nerve-muscle connections, thus emphasizing their plausible role to ensure the success of muscle regeneration. This review article aims to discuss the impact of aging on skeletal muscle regeneration and NMJs remodeling and will highlight the most recent insights about the role of semaphorins in this context.
Collapse
Affiliation(s)
- Damon Fard
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
| | - Alessandra Barbiera
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
| | - Gabriella Dobrowolny
- DAHFMO-Unità di Istologia ed Embriologia Medica, Sapienza Università di Roma, 00161 Roma, Italy.
| | - Luca Tamagnone
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| | - Bianca Maria Scicchitano
- Sezione di Istologia ed Embriologia, Dipartimento di Scienze della Vita e Sanità Pubblica,Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy.
| |
Collapse
|
13
|
Sui H, Dou J, Shi B, Cheng X. The reciprocity of skeletal muscle and bone: an evolving view from mechanical coupling, secretory crosstalk to stem cell exchange. Front Physiol 2024; 15:1349253. [PMID: 38505709 PMCID: PMC10949226 DOI: 10.3389/fphys.2024.1349253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/19/2024] [Indexed: 03/21/2024] Open
Abstract
Introduction: Muscle and bone constitute the two main parts of the musculoskeletal system and generate an intricately coordinated motion system. The crosstalk between muscle and bone has been under investigation, leading to revolutionary perspectives in recent years. Method and results: In this review, the evolving concept of muscle-bone interaction from mechanical coupling, secretory crosstalk to stem cell exchange was explained in sequence. The theory of mechanical coupling stems from the observation that the development and maintenance of bone mass are largely dependent on muscle-derived mechanical loads, which was later proved by Wolff's law, Utah paradigm and Mechanostat hypothesis. Then bone and muscle are gradually recognized as endocrine organs, which can secrete various cytokines to modulate the tissue homeostasis and remodeling to each other. The latest view presented muscle-bone interaction in a more direct way: the resident mesenchymal stromal cell in the skeletal muscle, i.e., fibro-adipogenic progenitors (FAPs), could migrate to the bone injury site and contribute to bone regeneration. Emerging evidence even reveals the ectopic source of FAPs from tissue outside the musculoskeletal system, highlighting its dynamic property. Conclusion: FAPs have been established as the critical cell connecting muscle and bone, which provides a new modality to study inter-tissue communication. A comprehensive and integrated perspective of muscle and bone will facilitate in-depth research in the musculoskeletal system and promote novel therapeutic avenues in treating musculoskeletal disorders.
Collapse
Affiliation(s)
| | | | | | - Xu Cheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Nielsen SDH, Sahebekhtiari N, Huang Z, Young JF, Rasmussen MK. Comparison of secreted miRNAs and proteins during proliferation and differentiation of bovine satellite cells in culture implies potential roles in regulating myogenesis. Gene 2024; 894:147979. [PMID: 37952749 DOI: 10.1016/j.gene.2023.147979] [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: 09/01/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Cultivated meat is an emerging new technology to produce sustainable meat for the future. The common approach for cultivated meat, is the isolation of satellite cells from donor animals, followed by in vitro proliferation and differentiation into primitive muscle fibers. The transformation of satellite cells into myofibers is tightly orchestrated by intra-cellular signaling, while the inter-cellular signaling is less well understood. Thus, the current study was conducted to map the secretion of potential signaling molecules (MicroRNAs and proteins) during proliferation and differentiation. Primary cultures of satellite cells were grown to 50% and 80% confluence, representing the proliferative phase or serum-starved for 1 and 3 days to induce differentiation. Post incubation in FBS-free media, the media were collected and analyzed for miRNA and protein content using gene-arrays and LC-MS/MS, respectively. When comparing the miRNA secretome at 50% and 80% confluence, we observed four differentially expressed miRNA, while only five were differentially expressed when comparing Day 1 to Day 3. A subsequent in silico analysis suggested that pathways of importance for myogenesis, e.g., MAPK and AMPK signaling, could be regulated by the secreted miRNAs. In addition, >300 proteins were secreted, including insulin-like growth factor 1 binding proteins 2, 3, 4, 5 and 6. In conclusion, this study demonstrated differential secretion of several miRNAs and proteins during both proliferation and differentiation of bovine satellite cells in vitro.
Collapse
Affiliation(s)
| | - Navid Sahebekhtiari
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | - Ziyu Huang
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | - Jette Feveile Young
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | | |
Collapse
|
15
|
Lee M, Park S, Choi B, Choi W, Lee H, Lee JM, Lee ST, Yoo KH, Han D, Bang G, Hwang H, Koh WG, Lee S, Hong J. Cultured meat with enriched organoleptic properties by regulating cell differentiation. Nat Commun 2024; 15:77. [PMID: 38167486 PMCID: PMC10762223 DOI: 10.1038/s41467-023-44359-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Research on cultured meat has primarily focused on the mass proliferation or differentiation of muscle cells; thus, the food characteristics of cultured meat remain relatively underexplored. As the quality of meat is determined by its organoleptic properties, cultured meat with similar sensory characteristics to animal-derived meat is highly desirable. In this study, we control the organoleptic and nutritional properties of cultured meat by tailoring the 2D differentiation of primary bovine myoblasts and primary bovine adipose-derived mesenchymal stem cells on gelatin/alginate scaffolds with varying stiffness. We assess the effect of muscle and adipose differentiation quality on the sensory properties of cultured meat. Thereafter, we fabricate cultured meat with similar sensory profiles to that of conventional beef by assembling the muscle and adipose constructs composed of highly differentiated cells. We introduce a strategy to produce cultured meat with enriched food characteristics by regulating cell differentiation with scaffold engineering.
Collapse
Affiliation(s)
- Milae Lee
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sohyeon Park
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Bumgyu Choi
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Woojin Choi
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Lee
- Department of Animal Life Science, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Jeong Min Lee
- Department of Applied Animal Life Science, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Seung Tae Lee
- Department of Animal Life Science, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
- Department of Applied Animal Life Science, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Ki Hyun Yoo
- Simple Planet, 805, 34, sangwan 12-gil, Seongdong-gu, Seoul, 04790, Republic of Korea
| | - Dongoh Han
- Simple Planet, 805, 34, sangwan 12-gil, Seongdong-gu, Seoul, 04790, Republic of Korea
| | - Geul Bang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Heeyoun Hwang
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sangmin Lee
- School of Mechanical Engineering, Chung-ang University, 84, Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Jinkee Hong
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
16
|
Lisco G, Disoteo OE, De Tullio A, De Geronimo V, Giagulli VA, Monzani F, Jirillo E, Cozzi R, Guastamacchia E, De Pergola G, Triggiani V. Sarcopenia and Diabetes: A Detrimental Liaison of Advancing Age. Nutrients 2023; 16:63. [PMID: 38201893 PMCID: PMC10780932 DOI: 10.3390/nu16010063] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Sarcopenia is an age-related clinical complaint characterized by the progressive deterioration of skeletal muscle mass and strength over time. Type 2 diabetes (T2D) is associated with faster and more relevant skeletal muscle impairment. Both conditions influence each other, leading to negative consequences on glycemic control, cardiovascular risk, general health status, risk of falls, frailty, overall quality of life, and mortality. PubMed/Medline, Scopus, Web of Science, and Google Scholar were searched for research articles, scientific reports, observational studies, clinical trials, narrative and systematic reviews, and meta-analyses to review the evidence on the pathophysiology of di-abetes-induced sarcopenia, its relevance in terms of glucose control and diabetes-related outcomes, and diagnostic and therapeutic challenges. The review comprehensively addresses key elements for the clinical definition and diagnostic criteria of sarcopenia, the pathophysiological correlation be-tween T2D, sarcopenia, and related outcomes, a critical review of the role of antihyperglycemic treatment on skeletal muscle health, and perspectives on the role of specific treatment targeting myokine signaling pathways involved in glucose control and the regulation of skeletal muscle metabolism and trophism. Prompt diagnosis and adequate management, including lifestyle inter-vention, health diet programs, micronutrient supplementation, physical exercise, and pharmaco-logical treatment, are needed to prevent or delay skeletal muscle deterioration in T2D.
Collapse
Affiliation(s)
- Giuseppe Lisco
- Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.T.); (V.A.G.); (E.J.); (E.G.)
| | - Olga Eugenia Disoteo
- Unit of Endocrinology, Diabetology, Dietetics and Clinical Nutrition, Sant Anna Hospital, 22020 San Fermo della Battaglia, Italy;
| | - Anna De Tullio
- Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.T.); (V.A.G.); (E.J.); (E.G.)
| | - Vincenzo De Geronimo
- Unit of Endocrinology, Clinical Diagnostic Center Morgagni, 95100 Catania, Italy;
| | - Vito Angelo Giagulli
- Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.T.); (V.A.G.); (E.J.); (E.G.)
| | - Fabio Monzani
- Geriatrics Unit, Department of Clinical & Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Emilio Jirillo
- Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.T.); (V.A.G.); (E.J.); (E.G.)
| | - Renato Cozzi
- Division of Endocrinology, Niguarda Hospital, 20162 Milan, Italy;
| | - Edoardo Guastamacchia
- Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.T.); (V.A.G.); (E.J.); (E.G.)
| | - Giovanni De Pergola
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy;
| | - Vincenzo Triggiani
- Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases, Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.T.); (V.A.G.); (E.J.); (E.G.)
| |
Collapse
|
17
|
Stella R, Bonadio RS, Cagnin S, Andreotti R, Massimino ML, Bertoli A, Peggion C. Secreted Metabolome of ALS-Related hSOD1(G93A) Primary Cultures of Myocytes and Implications for Myogenesis. Cells 2023; 12:2751. [PMID: 38067180 PMCID: PMC10706027 DOI: 10.3390/cells12232751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a motor neuron (MN) disease associated with progressive muscle atrophy, paralysis, and eventually death. Growing evidence demonstrates that the pathological process leading to ALS is the result of multiple altered mechanisms occurring not only in MNs but also in other cell types inside and outside the central nervous system. In this context, the involvement of skeletal muscle has been the subject of a few studies on patients and ALS animal models. In this work, by using primary myocytes derived from the ALS transgenic hSOD1(G93A) mouse model, we observed that the myogenic capability of such cells was defective compared to cells derived from control mice expressing the nonpathogenic hSOD1(WT) isoform. The correct in vitro myogenesis of hSOD1(G93A) primary skeletal muscle cells was rescued by the addition of a conditioned medium from healthy hSOD1(WT) myocytes, suggesting the existence of an in trans activity of secreted factors. To define a dataset of molecules participating in such safeguard action, we conducted comparative metabolomic profiling of a culture medium collected from hSOD1(G93A) and hSOD1(WT) primary myocytes and report here an altered secretion of amino acids and lipid-based signaling molecules. These findings support the urgency of better understanding the role of the skeletal muscle secretome in the regulation of the myogenic program and mechanisms of ALS pathogenesis and progression.
Collapse
Affiliation(s)
- Roberto Stella
- Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy
| | | | - Stefano Cagnin
- Department of Biology, University of Padova, 35131 Padova, Italy (S.C.)
- CIR-Myo Myology Center, University of Padova, 35131 Padova, Italy
| | - Roberta Andreotti
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy (A.B.)
| | - Maria Lina Massimino
- Neuroscience Institute, Consiglio Nazionale delle Ricerche, 35131 Padova, Italy;
| | - Alessandro Bertoli
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy (A.B.)
- Neuroscience Institute, Consiglio Nazionale delle Ricerche, 35131 Padova, Italy;
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
| | - Caterina Peggion
- Department of Biology, University of Padova, 35131 Padova, Italy (S.C.)
| |
Collapse
|
18
|
Hota M, Barber JL, Ruiz-Ramie JJ, Schwartz CS, Lam DTUH, Rao P, Mi MY, Katz DH, Robbins JM, Clish CB, Gerszten RE, Sarzynski MA, Ghosh S, Bouchard C. Omics-driven investigation of the biology underlying intrinsic submaximal working capacity and its trainability. Physiol Genomics 2023; 55:517-543. [PMID: 37661925 PMCID: PMC11178266 DOI: 10.1152/physiolgenomics.00163.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/21/2023] [Accepted: 08/31/2023] [Indexed: 09/05/2023] Open
Abstract
Submaximal exercise capacity is an indicator of cardiorespiratory fitness with clinical and public health implications. Submaximal exercise capacity and its response to exercise programs are characterized by heritability levels of about 40%. Using physical working capacity (power output) at a heart rate of 150 beats/min (PWC150) as an indicator of submaximal exercise capacity in subjects of the HERITAGE Family Study, we have undertaken multi-omics and in silico explorations of the underlying biology of PWC150 and its response to 20 wk of endurance training. Our goal was to illuminate the biological processes and identify panels of genes associated with human variability in intrinsic PWC150 (iPWC150) and its trainability (dPWC150). Our bioinformatics approach was based on a combination of genome-wide association, skeletal muscle gene expression, and plasma proteomics and metabolomics experiments. Genes, proteins, and metabolites showing significant associations with iPWC150 or dPWC150 were further queried for the enrichment of biological pathways. We compared genotype-phenotype associations of emerging candidate genes with reported functional consequences of gene knockouts in mouse models. We investigated the associations between DNA variants and multiple muscle and cardiovascular phenotypes measured in HERITAGE subjects. Two panels of prioritized genes of biological relevance to iPWC150 (13 genes) and dPWC150 (6 genes) were identified, supporting the hypothesis that genes and pathways associated with iPWC150 are different from those underlying dPWC150. Finally, the functions of these genes and pathways suggested that human variation in submaximal exercise capacity is mainly driven by skeletal muscle morphology and metabolism and red blood cell oxygen-carrying capacity.NEW & NOTEWORTHY Multi-omics and in silico explorations of the genes and underlying biology of submaximal exercise capacity and its response to 20 wk of endurance training were undertaken. Prioritized genes were identified: 13 genes for variation in submaximal exercise capacity in the sedentary state and 5 genes for the response level to endurance training, with no overlap between them. Genes and pathways associated with submaximal exercise capacity in the sedentary state are different from those underlying trainability.
Collapse
Affiliation(s)
- Monalisa Hota
- Centre for Computational Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Jacob L Barber
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
| | - Jonathan J Ruiz-Ramie
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
- Department of Kinesiology, Augusta University, Augusta, Georgia, United States
| | - Charles S Schwartz
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
| | - Do Thuy Uyen Ha Lam
- Centre for Computational Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Prashant Rao
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Michael Y Mi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Daniel H Katz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Jeremy M Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Clary B Clish
- Metabolomics Platform, Broad Institute, Boston, Massachusetts, United States
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| | - Mark A Sarzynski
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, United States
| | - Sujoy Ghosh
- Centre for Computational Biology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Bioinformatics Section, Human Genomics Core, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
- Program in Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, United States
| |
Collapse
|
19
|
Hanson B, Vorobieva I, Zheng W, Conceição M, Lomonosova Y, Mäger I, Puri PL, El Andaloussi S, Wood MJ, Roberts TC. EV-mediated promotion of myogenic differentiation is dependent on dose, collection medium, and isolation method. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:511-528. [PMID: 37602275 PMCID: PMC10432918 DOI: 10.1016/j.omtn.2023.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 07/11/2023] [Indexed: 08/22/2023]
Abstract
Extracellular vesicles (EVs) have been implicated in the regulation of myogenic differentiation. C2C12 murine myoblast differentiation was reduced following treatment with GW4869 or heparin (to inhibit exosome biogenesis and EV uptake, respectively). Conversely, treatment with C2C12 myotube-conditioned medium enhanced myogenic differentiation. Ultrafiltration-size exclusion liquid chromatography (UF-SEC) was used to isolate EVs and non-EV extracellular protein in parallel from C2C12 myoblast- and myotube-conditioned medium. UF-SEC-purified EVs promoted myogenic differentiation at low doses (≤2 × 108 particles/mL) and were inhibitory at the highest dose tested (2 × 1011 particles/mL). Conversely, extracellular protein fractions had no effect on myogenic differentiation. While the transfer of muscle-enriched miRNAs (myomiRs) has been proposed to mediate the pro-myogenic effects of EVs, we observed that they are scarce in EVs (e.g., 1 copy of miR-133a-3p per 195 EVs). Furthermore, we observed pro-myogenic effects with undifferentiated myoblast-derived EVs, in which myomiR concentrations are even lower, suggestive of a myomiR-independent mechanism underlying the observed pro-myogenic effects. During these investigations we identified technical factors with profound confounding effects on myogenic differentiation. Specifically, co-purification of insulin (a component of Opti-MEM) in non-EV LC fractions and polymer precipitated EV preparations. These findings provide further evidence that polymer-based precipitation techniques should be avoided in EV research.
Collapse
Affiliation(s)
- Britt Hanson
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Ioulia Vorobieva
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Wenyi Zheng
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge SE-141 86, Sweden
| | - Mariana Conceição
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Yulia Lomonosova
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Imre Mäger
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Pier Lorenzo Puri
- Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA 92037, USA
| | - Samir El Andaloussi
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge SE-141 86, Sweden
| | - Matthew J.A. Wood
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX3 7TY, UK
| | - Thomas C. Roberts
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX3 7TY, UK
| |
Collapse
|
20
|
Alheib O, da Silva LP, Mesquita KA, da Silva Morais A, Pirraco RP, Reis RL, Correlo VM. Human adipose-derived mesenchymal stem cells laden in gellan gum spongy-like hydrogels for volumetric muscle loss treatment. Biomed Mater 2023; 18:065005. [PMID: 37604159 DOI: 10.1088/1748-605x/acf25b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/21/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND volumetric muscle loss (VML) is a traumatic massive loss of muscular tissue which frequently leads to amputation, limb loss, or lifetime disability. The current medical intervention is limited to autologous tissue transfer, which usually leads to non-functional tissue recovery. Tissue engineering holds a huge promise for functional recovery. METHODS in this work, we evaluated the potential of human adipose-derived mesenchymal stem cells (hASCs) pre-cultured in gellan gum based spongy-like hydrogels (SLHs). RESULTS in vitro, hASCs were spreading, proliferating, and releasing growth factors and cytokines (i.e. fibroblast growth factor, hepatocyte growth factor, insulin-like growth factor 1, interleukin-6 (IL-6), IL-8, IL-10, vascular endothelial growth factor) important for muscular regeneration. After implantation into a volumetric muscle loss (VML) mouse model, implants were degrading overtime, entirely integrating into the host between 4 and 8 weeks. In both SLH and SLH + hASCs defects, infiltrated cells were observed inside constructs associated with matrix deposition. Also, minimal collagen deposition was marginally observed around the constructs along both time-points. Neovascularization (CD31+vessels) and neoinnervation (β-III tubulin+bundles) were significantly detected in the SLH + hASCs group, in relation to the SHAM (empty lesion). A higher density ofα-SA+and MYH7+cells were found in the injury site among all different experimental groups, at both time-points, in relation to the SHAM. The levels ofα-SA, MyoD1, and myosin heavy chain proteins were moderately increased in the SLH + hASCs group after 4 weeks, and in the hASCs group after 8 weeks, in relation to the SHAM. CONCLUSIONS taken together, defects treated with hASCs-laden SLH promoted angiogenesis, neoinnervation, and the expression of myogenic proteins.
Collapse
Affiliation(s)
- Omar Alheib
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lucilia P da Silva
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Katia A Mesquita
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alain da Silva Morais
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Vitor M Correlo
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
21
|
Montgomery MK, De Nardo W, Watt MJ. Exercise training induces depot-specific remodeling of protein secretion in skeletal muscle and adipose tissue of obese male mice. Am J Physiol Endocrinol Metab 2023; 325:E227-E238. [PMID: 37493472 DOI: 10.1152/ajpendo.00178.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
Acute exercise induces changes in circulating proteins, which are known to alter metabolism and systemic energy balance. Skeletal muscle is a primary contributor to changes in the plasma proteome with acute exercise. An important consideration when assessing the endocrine function of muscle is the presence of different fiber types, which show distinct functional and metabolic properties and likely secrete different proteins. Similarly, adipokines are important regulators of systemic metabolism and have been shown to differ between depots. Given the health-promoting effects of exercise, we proposed that understanding depot-specific remodeling of protein secretion in muscle and adipose tissue would provide new insights into intertissue communication and uncover novel regulators of energy homeostasis. Here, we examined the effect of endurance exercise training on protein secretion from fast-twitch extensor digitorum longus (EDL) and slow-twitch soleus muscle and visceral and subcutaneous adipose tissue. High-fat diet-fed mice were exercise trained for 6 wk, whereas a Control group remained sedentary. Secreted proteins from excised EDL and soleus muscle, inguinal, and epididymal adipose tissues were detected using mass spectrometry. We detected 575 and 784 secreted proteins from EDL and soleus muscle and 738 and 920 proteins from inguinal and epididymal adipose tissue, respectively. Of these, 331 proteins were secreted from all tissues, whereas secretion of many other proteins was tissue and depot specific. Exercise training led to substantial remodeling of protein secretion from EDL, whereas soleus showed only minor changes. Myokines released exclusively from EDL or soleus were associated with glycogen metabolism and cellular stress response, respectively. Adipokine secretion was completely refractory to exercise regulation in both adipose depots. This study provides an in-depth resource of protein secretion from muscle and adipose tissue, and its regulation following exercise training, and identifies distinct depot-specific secretion patterns that are related to the metabolic properties of the tissue of origin.NEW & NOTEWORTHY The present study examines the effects of exercise training on protein secretion from fast-twitch and slow-twitch muscle as well as visceral and subcutaneous adipose tissue of obese mice. Although exercise training leads to substantial remodeling of protein secretion from fast-twitch muscle, adipose tissue is completely refractory to exercise regulation.
Collapse
Affiliation(s)
- Magdalene K Montgomery
- Faculty of Medicine, Dentistry & Health Sciences, Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - William De Nardo
- Faculty of Medicine, Dentistry & Health Sciences, Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Matthew J Watt
- Faculty of Medicine, Dentistry & Health Sciences, Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
22
|
Maeno T, Arimatsu R, Ojima K, Yamaya Y, Imakyure H, Watanabe N, Komiya Y, Kobayashi K, Nakamura M, Nishimura T, Tatsumi R, Suzuki T. Netrin-4 synthesized in satellite cell-derived myoblasts stimulates autonomous fusion. Exp Cell Res 2023; 430:113698. [PMID: 37437770 DOI: 10.1016/j.yexcr.2023.113698] [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: 12/08/2022] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023]
Abstract
Satellite cells are indispensable for skeletal muscle regeneration and hypertrophy by forming nascent myofibers (myotubes). They synthesize multi-potent modulator netrins (secreted subtypes: netrin-1, -3, and -4), originally found as classical neural axon guidance molecules. While netrin-1 and -3 have key roles in myogenic differentiation, the physiological significance of netrin-4 is still unclear. This study examined whether netrin-4 regulates myofiber type commitment and myotube formation. Initially, the expression profiles indicated that satellite cells isolated from the extensor digitorum longus muscle (EDL muscle: fast-twitch myofiber-abundant) expressed slightly more netrin-4 than the soleus muscle (slow-type abundant) cells. As netrin-4 knockdown inhibited both slow- and fast-type myotube formation, netrin-4 may not directly regulate myofiber type commitment. However, netrin-4 knockdown in satellite cell-derived myoblasts reduced the myotube fusion index, while exogenous netrin-4 promoted myotube formation, even though netrin-4 expression level was maximum during the initiation stage of myogenic differentiation. Furthermore, netrin-4 knockdown also inhibited MyoD (a master transcriptional factor of myogenesis) and Myomixer (a myoblast fusogenic molecule) expression. These data suggest that satellite cells synthesize netrin-4 during myogenic differentiation initiation to promote their own fusion, stimulating the MyoD-Myomixer signaling axis.
Collapse
Affiliation(s)
- Takahiro Maeno
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Rio Arimatsu
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Koichi Ojima
- Muscle Biology Research Unit, Division of Animal Products Research, Institute of Livestock and Grassland Science, NARO, Tsukuba, Japan
| | - Yuki Yamaya
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Hikaru Imakyure
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Naruha Watanabe
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Yusuke Komiya
- Department of Animal Science, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Mako Nakamura
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Graduate School of Agriculture, Hokkaido University, Sapporo, Japan
| | - Ryuichi Tatsumi
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan
| | - Takahiro Suzuki
- Laboratory of Muscle and Meat Science, Department of Animal and Marine Bioresource Sciences, Research Faculty of Agriculture, Graduate School of Agriculture, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
23
|
Geladari E, Alexopoulos T, Kontogianni MD, Vasilieva L, Mani I, Alexopoulou A. Mechanisms of sarcopenia in liver cirrhosis and the role of myokines. Ann Gastroenterol 2023; 36:392-404. [PMID: 37396001 PMCID: PMC10304523 DOI: 10.20524/aog.2023.0804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 04/19/2023] [Indexed: 07/04/2023] Open
Abstract
Sarcopenia is a syndrome characterized by a decline in skeletal muscle quantity and/or quality, strength and performance, leading to unfortunate events, such as injurious falls or even death. It is not identical to frailty and malnutrition, even though there is a significant overlap among these syndromes. In patients with liver cirrhosis (LC), sarcopenia is classified as secondary and has been associated with increased morbidity and mortality during the pre- and post-transplantation period. It can be a result of malnutrition, hyperammonemia, low physical activity, endocrine abnormalities, accelerated starvation, metabolic disturbances, altered gut function leading to chronic inflammation, and alcohol abuse. Myokines are peptides mainly synthesized by contracting muscle and adipose tissue cells and may play a key role in the pathophysiology of sarcopenia. More than a hundred myokines have been recognized, but only a few have been investigated. They can be classified as negative regulators, such as myostatin, tumor growth factor-β, activins, growth differentiation factor-11, and positive regulators of muscle growth including follistatin, bone morphogenic proteins, and irisin. So far, only myostatin, follistatin, irisin and decorin have been studied in LC-associated sarcopenia. In this review, we focused on the mechanisms of cirrhosis-related sarcopenia and the role of myokines that have already been studied in the literature, either as markers helping in the diagnostic evaluation of sarcopenia, or as prognostic factors of survival. Standard therapeutic options to prevent or treat sarcopenia in LC are also being reported, as well as the possible therapeutic implication of myokines.
Collapse
Affiliation(s)
- Eleni Geladari
- 2 Department of Internal Medicine and Research Laboratory, Medical School, National & Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece (Eleni Geladari, Theodoros Alexopoulos, Iliana Mani, Alexandra Alexopoulou)
| | - Theodoros Alexopoulos
- 2 Department of Internal Medicine and Research Laboratory, Medical School, National & Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece (Eleni Geladari, Theodoros Alexopoulos, Iliana Mani, Alexandra Alexopoulou)
| | - Meropi D. Kontogianni
- Department of Nutrition and Dietetics, School of Health Science & Education, Harokopio University, Athens, Greece (Meropi D. Kontogianni)
| | - Larisa Vasilieva
- Gastroenterology Department, Alexandra Hospital (Larisa Vasilieva), Athens, Greece
| | - Iliana Mani
- 2 Department of Internal Medicine and Research Laboratory, Medical School, National & Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece (Eleni Geladari, Theodoros Alexopoulos, Iliana Mani, Alexandra Alexopoulou)
| | - Alexandra Alexopoulou
- 2 Department of Internal Medicine and Research Laboratory, Medical School, National & Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece (Eleni Geladari, Theodoros Alexopoulos, Iliana Mani, Alexandra Alexopoulou)
| |
Collapse
|
24
|
Feng Y, Cui Z, Lu X, Gong H, Liu X, Wang H, Cheng H, Gao H, Shi X, Li Y, Ye H, Zhang Q, Kong X. Transcriptomics Dissection of Calorie Restriction and Exercise Training in Brown Adipose Tissue and Skeletal Muscle. Nutrients 2023; 15:nu15041047. [PMID: 36839405 PMCID: PMC9966723 DOI: 10.3390/nu15041047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Calorie restriction (CR) and exercise training (EX) are two critical lifestyle interventions for the prevention and treatment of metabolic diseases, such as obesity and diabetes. Brown adipose tissue (BAT) and skeletal muscle are two important organs for the generation of heat. Here, we undertook detailed transcriptional profiling of these two thermogenic tissues from mice treated subjected to CR and/or EX. We found transcriptional reprogramming of BAT and skeletal muscle as a result of CR but little from EX. Consistent with this, CR induced alterations in the expression of genes encoding adipokines and myokines in BAT and skeletal muscle, respectively. Deconvolution analysis showed differences in the subpopulations of myogenic cells, mesothelial cells and endogenic cells in BAT and in the subpopulations of satellite cells, immune cells and endothelial cells in skeletal muscle as a result of CR or EX. NicheNet analysis, exploring potential inter-organ communication, indicated that BAT and skeletal muscle could mutually regulate their fatty acid metabolism and thermogenesis through ligands and receptors. These data comprise an extensive resource for the study of thermogenic tissue molecular responses to CR and/or EX in a healthy state.
Collapse
Affiliation(s)
- Yonghao Feng
- Department of Endocrinology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Zhicheng Cui
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaodan Lu
- Precision Medicine Center, Jilin Province General Hospital, Changchun 130021, China
| | - Hongyu Gong
- School of Life Sciences, Inner Mongolia University, Hohhot 010000, China
| | - Xiaoyu Liu
- School of Life Sciences, Inner Mongolia University, Hohhot 010000, China
| | - Hui Wang
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Haoyu Cheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Huanqing Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaohong Shi
- Department of Endocrinology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qiongyue Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Correspondence: (Q.Z.); (X.K.)
| | - Xingxing Kong
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Huashan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China
- Correspondence: (Q.Z.); (X.K.)
| |
Collapse
|
25
|
Vanhorebeek I, Gunst J, Casaer MP, Derese I, Derde S, Pauwels L, Segers J, Hermans G, Gosselink R, Van den Berghe G. Skeletal Muscle Myokine Expression in Critical Illness, Association With Outcome and Impact of Therapeutic Interventions. J Endocr Soc 2023; 7:bvad001. [PMID: 36726836 PMCID: PMC9879715 DOI: 10.1210/jendso/bvad001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Indexed: 01/09/2023] Open
Abstract
Context Muscle expresses and secretes several myokines that bring about benefits in distant organs. Objective We investigated the impact of critical illness on muscular expression of irisin, kynurenine aminotransferases, and amylase; association with clinical outcome; and impact of interventions that attenuate muscle wasting/weakness. Methods We studied critically ill patients who participated in 2 randomized controlled trials (EPaNIC/NESCI) and documented time profiles in critically ill mice. Included in the study were 174 intensive care unit (ICU) patients (day 8 ± 1) vs 19 matched controls, and 60 mice subjected to surgery/sepsis vs 60 pair-fed healthy mice. Interventions studied included 7-day neuromuscular electrical stimulation (NMES), and withholding parenteral nutrition (PN) in the first ICU week (late PN) vs early PN. The main outcome measures were FNDC5 (irisin- precursor), KYAT1, KYAT3, and amylase mRNA expression in skeletal muscle. Results Critically ill patients showed 34% to 80% lower mRNA expression of FNDC5, KYAT1, and amylases than controls (P < .0001). Critically ill mice showed time-dependent reductions in all mRNAs compared with healthy mice (P ≤ .04). The lower FNDC5 expression in patients was independently associated with a higher ICU mortality (P = .015) and ICU-acquired weakness (P = .012), whereas the lower amylase expression in ICU survivors was independently associated with a longer ICU stay (P = .0060). Lower amylase expression was independently associated with a lower risk of death (P = .048), and lower KYAT1 expression with a lower risk of weakness (P = .022). NMES increased FNDC5 expression compared with unstimulated muscle (P = .016), and late PN patients had a higher KYAT1 expression than early PN patients (P = .022). Conclusion Expression of the studied myokines was affected by critical illness and associated with clinical outcomes, with limited effects of interventions that attenuate muscle wasting or weakness.
Collapse
Affiliation(s)
- Ilse Vanhorebeek
- Correspondence: Prof. Ilse Vanhorebeek, MEng, PhD, Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;. ; or Prof. Greet Van den Berghe, MD, PhD, Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | - Jan Gunst
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium,Clinical Division of Intensive Care Medicine, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Michaël P Casaer
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium,Clinical Division of Intensive Care Medicine, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Inge Derese
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Sarah Derde
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Lies Pauwels
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Johan Segers
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Greet Hermans
- Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium,Medical Intensive Care Unit, Department of General Internal Medicine, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Rik Gosselink
- Department of Rehabilitation Sciences, Faculty of Kinesiology and Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Greet Van den Berghe
- Correspondence: Prof. Ilse Vanhorebeek, MEng, PhD, Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;. ; or Prof. Greet Van den Berghe, MD, PhD, Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
26
|
Chodkowska KA, Wódz K, Wojciechowski J. Sustainable Future Protein Foods: The Challenges and the Future of Cultivated Meat. Foods 2022; 11:foods11244008. [PMID: 36553750 PMCID: PMC9778282 DOI: 10.3390/foods11244008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/19/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Global pressure from consumers to improve animal welfare, and reduce microbiological risks or the use of antibiotics pose new challenges for the meat industry. Today's livestock production, despite many undertaken measures, is still far from being sustainable. This forced the need to work on alternative protein types that come from plants, insects, fungi, or cell culture processes. Due to some technical and legal barriers, cultivated meat is not present on the European market, however, in 2020 it was approved in Singapore and in 2022 in the USA. While the technology of obtaining cell cultures from animal muscles has been known and successfully practiced for years, the production of a stable piece of meat with appropriate texture, taste, and smell, is still a problem for several scientific groups related to subsequent companies trying to obtain the highest quality product, in line with the expectations of customers. Although the work on optimal cell meat production has been going on for years, it is still in an early stage, mainly due to several limitations that represent milestones for industrial production. The most important are: the culture media (without animal serum), which will provide an environment for optimal muscle development, natural or close to natural (but still safe for the consumer) stable scaffolds for growing cells. Here, we review the actual knowledge about the above-mentioned challenges which make the production of cellular meat not yet developed on an industrial scale.
Collapse
Affiliation(s)
| | - Karolina Wódz
- Laboratory of Molecular Biology, Vet-Lab Brudzew, Turkowska 58c, 62-720 Brudzew, Poland
| | - Jakub Wojciechowski
- Laboratory of Molecular Biology, Vet-Lab Brudzew, Turkowska 58c, 62-720 Brudzew, Poland
| |
Collapse
|
27
|
Yedigaryan L, Martínez-Sarrà E, Giacomazzi G, Giarratana N, van der Veer BK, Rotini A, Querceto S, Grosemans H, Cortés-Calabuig Á, Salucci S, Battistelli M, Falcieri E, Gijsbers R, Quattrocelli M, Peng Koh K, De Waele L, Buyse GM, Derua R, Sampaolesi M. Extracellular vesicle-derived miRNAs improve stem cell-based therapeutic approaches in muscle wasting conditions. Front Immunol 2022; 13:977617. [PMID: 36451814 PMCID: PMC9702803 DOI: 10.3389/fimmu.2022.977617] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/17/2022] [Indexed: 11/15/2022] Open
Abstract
Skeletal muscle holds an intrinsic capability of growth and regeneration both in physiological conditions and in case of injury. Chronic muscle illnesses, generally caused by genetic and acquired factors, lead to deconditioning of the skeletal muscle structure and function, and are associated with a significant loss in muscle mass. At the same time, progressive muscle wasting is a hallmark of aging. Given the paracrine properties of myogenic stem cells, extracellular vesicle-derived signals have been studied for their potential implication in both the pathogenesis of degenerative neuromuscular diseases and as a possible therapeutic target. In this study, we screened the content of extracellular vesicles from animal models of muscle hypertrophy and muscle wasting associated with chronic disease and aging. Analysis of the transcriptome, protein cargo, and microRNAs (miRNAs) allowed us to identify a hypertrophic miRNA signature amenable for targeting muscle wasting, consisting of miR-1 and miR-208a. We tested this signature among others in vitro on mesoangioblasts (MABs), vessel-associated adult stem cells, and we observed an increase in the efficiency of myogenic differentiation. Furthermore, injections of miRNA-treated MABs in aged mice resulted in an improvement in skeletal muscle features, such as muscle weight, strength, cross-sectional area, and fibrosis compared to controls. Overall, we provide evidence that the extracellular vesicle-derived miRNA signature we identified enhances the myogenic potential of myogenic stem cells.
Collapse
Affiliation(s)
- Laura Yedigaryan
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Ester Martínez-Sarrà
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Giorgia Giacomazzi
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Nefele Giarratana
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Bernard K. van der Veer
- Department of Development and Regeneration, Laboratory for Stem Cell and Developmental Epigenetics, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Alessio Rotini
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Silvia Querceto
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Hanne Grosemans
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Álvaro Cortés-Calabuig
- Laboratory for Cytogenetics and Genome Research, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Sara Salucci
- Cellular Signalling Laboratory, Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, Urbino University Carlo Bo, Urbino, Italy
| | - Elisabetta Falcieri
- Department of Biomolecular Sciences, Urbino University Carlo Bo, Urbino, Italy
| | - Rik Gijsbers
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - Mattia Quattrocelli
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium,Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, Heart Institute, University of Cincinnati College of Medicine and Molecular Cardiovascular Biology Division, Cincinnati, OH, United States
| | - Kian Peng Koh
- Department of Development and Regeneration, Laboratory for Stem Cell and Developmental Epigenetics, Stem Cell Institute, KU Leuven, Leuven, Belgium
| | - Liesbeth De Waele
- Department of Development and Regeneration, Pediatric Neurology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Gunnar M. Buyse
- Department of Development and Regeneration, Pediatric Neurology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, SyBioMa, KU Leuven, Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium,Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University of Rome, Rome, Italy,*Correspondence: Maurilio Sampaolesi,
| |
Collapse
|
28
|
Waldemer-Streyer RJ, Kim D, Chen J. Muscle cell-derived cytokines in skeletal muscle regeneration. FEBS J 2022; 289:6463-6483. [PMID: 35073461 PMCID: PMC9308828 DOI: 10.1111/febs.16372] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 12/14/2022]
Abstract
Regeneration of the mammalian adult skeletal muscle is a well-orchestrated process regulated by multiple proteins and signalling pathways. Cytokines constitute a major class of regulators of skeletal myogenesis. It is well established that infiltrating immune cells at the site of muscle injury secrete cytokines, which play critical roles in the myofibre repair and regeneration process. In the past 10-15 years, skeletal muscle itself has emerged as a prolific producer of cytokines. Much attention in the field has been focused on the endocrine effects of muscle-secreted cytokines (myokines) on metabolic regulation. However, ample evidence suggests that muscle-derived cytokines also regulate myogenic differentiation and muscle regeneration in an autocrine manner. In this review, we survey cytokines that meet two criteria: (a) evidence of expression by muscle cells; (b) evidence demonstrating a myogenic function. Dozens of cytokines representing several major classes make up this group, and together they regulate all steps of the myogenic process. How such a large array of cytokines coordinate their signalling to form a regulatory network is a fascinating, pressing question. Functional studies that can distinguish the source of the cytokines in vivo are also much needed in order to facilitate exploration of their full therapeutic potential.
Collapse
Affiliation(s)
| | | | - Jie Chen
- Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, 601 S. Goodwin Ave., Urbana, IL 61801
| |
Collapse
|
29
|
Zamboni M, Mazzali G, Brunelli A, Saatchi T, Urbani S, Giani A, Rossi AP, Zoico E, Fantin F. The Role of Crosstalk between Adipose Cells and Myocytes in the Pathogenesis of Sarcopenic Obesity in the Elderly. Cells 2022; 11:3361. [PMID: 36359757 PMCID: PMC9655977 DOI: 10.3390/cells11213361] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/08/2022] [Accepted: 10/14/2022] [Indexed: 11/15/2023] Open
Abstract
As a result of aging, body composition changes, with a decline in muscle mass and an increase in adipose tissue (AT), which reallocates from subcutaneous to visceral depots and stores ectopically in the liver, heart and muscles. Furthermore, with aging, muscle and AT, both of which have recognized endocrine activity, become dysfunctional and contribute, in the case of positive energy balance, to the development of sarcopenic obesity (SO). SO is defined as the co-existence of excess adiposity and low muscle mass and function, and its prevalence increases with age. SO is strongly associated with greater morbidity and mortality. The pathogenesis of SO is complex and multifactorial. This review focuses mainly on the role of crosstalk between age-related dysfunctional adipose and muscle cells as one of the mechanisms leading to SO. A better understanding of this mechanisms may be useful for development of prevention strategies and treatments aimed at reducing the occurrence of SO.
Collapse
Affiliation(s)
- Mauro Zamboni
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Gloria Mazzali
- Geriatrics Division, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Anna Brunelli
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Tanaz Saatchi
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Silvia Urbani
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Anna Giani
- Geriatrics Division, Department of Surgery, Dentistry, Pediatric and Gynecology, Healthy Aging Center, University of Verona, 37126 Verona, Italy
| | - Andrea P. Rossi
- Geriatrics Division, Department of Medicine, AULSS2, Ospedale Ca’Foncello, 31100 Treviso, Italy
| | - Elena Zoico
- Geriatrics Division, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Francesco Fantin
- Geriatrics Division, Department of Medicine, University of Verona, 37126 Verona, Italy
| |
Collapse
|
30
|
Abstract
Exosomes are a class of extracellular vesicles with a diameter of 50-100 nm secreted by various cells. They are generated through complex intracellular production mechanisms before being secreted to the extracellular environment. Due to their inclusion of proteins, lipids, and nucleic acids, exosomes play an important role in intercellular communication. Pancreatic β-cells play an irreplaceable role in the body's glucose metabolism. Their dysfunction is one of the causes of diabetes. Exosomes of various cells regulate the function of β-cells by regulating autoimmunity, delivering non-coding RNAs, or directly regulating intracellular signal pathways. This communication between β-cells and other cells plays an important role in the pathogenesis and development of diabetes, and has potential for clinical application. This paper reviews the biological sources and functions of exosomes, as well as intercellular crosstalk between β-cells and other cells that is involved in β-cell failure and regeneration.
Collapse
Affiliation(s)
- Yu Wu
- Diabetes Research Center, Medical School, Ningbo University, Ningbo, China
| | - Qin Huang
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Shizhong Bu
- Diabetes Research Center, Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
31
|
Gentile GM, Gamarra JR, Engels NM, Blue RE, Hoerr I, Wiedner HJ, Hinkle ER, Cote JL, Leverence E, Mills CA, Herring LE, Tan X, Giudice J. The synaptosome-associated protein 23 (SNAP23) is necessary for proper myogenesis. FASEB J 2022; 36:e22441. [PMID: 35816155 PMCID: PMC9836321 DOI: 10.1096/fj.202101627rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 01/14/2023]
Abstract
Vesicle-mediated transport is necessary for maintaining cellular homeostasis and proper signaling. The synaptosome-associated protein 23 (SNAP23) is a member of the SNARE protein family and mediates the vesicle docking and membrane fusion steps of secretion during exocytosis. Skeletal muscle has been established as a secretory organ; however, the role of SNAP23 in the context of skeletal muscle development is still unknown. Here, we show that depletion of SNAP23 in C2C12 mouse myoblasts reduces their ability to differentiate into myotubes as a result of premature cell cycle exit and early activation of the myogenic transcriptional program. This effect is rescued when cells are seeded at a high density or when cultured in conditioned medium from wild type cells. Proteomic analysis of collected medium indicates that SNAP23 depletion leads to a misregulation of exocytosis, including decreased secretion of the insulin-like growth factor 1 (IGF1), a critical protein for muscle growth, development, and function. We further demonstrate that treatment of SNAP23-depleted cells with exogenous IGF1 rescues their myogenic capacity. We propose that SNAP23 mediates the secretion of specific proteins, such as IGF1, that are important for achieving proper differentiation of skeletal muscle cells during myogenesis. This work highlights the underappreciated role of skeletal muscle as a secretory organ and contributes to the understanding of factors necessary for myogenesis.
Collapse
Affiliation(s)
- Gabrielle M. Gentile
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer R. Gamarra
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nichlas M. Engels
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - R. Eric Blue
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Isabel Hoerr
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hannah J. Wiedner
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Emma R. Hinkle
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica L. Cote
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elise Leverence
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christine A. Mills
- UNC Proteomics Core Facility, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Laura E. Herring
- UNC Proteomics Core Facility, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xianming Tan
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jimena Giudice
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
32
|
Wang J, Liu C, Zhang L, Liu N, Wang L, Wu J, Wang Y, Hao H, Cao L, Yuan S, Huang L. Prevalence and associated factors of possible sarcopenia and sarcopenia: findings from a Chinese community-dwelling old adults cross-sectional study. BMC Geriatr 2022; 22:592. [PMID: 35850661 PMCID: PMC9290196 DOI: 10.1186/s12877-022-03286-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/11/2022] [Indexed: 11/10/2022] Open
Abstract
Purpose To describe the prevalence and analyse the associated factors of possible sarcopenia and sarcopenia among community-dwelling old adults in China, in order to provide effective strategies for early prevention and treatment of sarcopenia. Methods This cross-sectional study evaluated community-dwelling old adults aged over 60 years. The basic information, morphological indices, body composition, physical activities were collected and assessed. Possible sarcopenia and sarcopenia were diagnosed by the criteria of Asian Working Group for Sarcopenia (AWGS) in 2019. A multivariate logistic regression model with stepwise method was employed to identify factors associated with possible sarcopenia and sarcopenia. Results In total 729 old adults from Tianjin were included in this study. Eighty-one participants were diagnosed with possible sarcopenia (prevalence of 11.11%). Seventy-five participants were diagnosed with sarcopenia (prevalence of 10.29%). Age (odds ratio (OR):1.047, 95% confidence interval (CI): 1.055–1.090) and lower physical activities (low level OR:4.171, 95% CI:1.790–9.720; medium level OR:2.634, 95% CI:1.352–5.132) were significantly associated with possible sarcopenia. Age (OR:1.187, 95% CI:1.124–1.253), higher body fat percentage (OR:1.225, 95% CI:1.140–1.317), lower BMI (OR:0.424, 95% CI:0.346–0.519), lower mini-mental state examination (MMSE) scores (OR:0.865,95% CI:0.781–0.958) and low physical activities (OR:4.638, 95% CI:1.683–12.782) were significantly associated with sarcopenia. Conclusion Possible sarcopenia and sarcopenia are prevalent among community-dwelling old adults in China. Ageing and lower physical activities were both associated with possible sarcopenia and sarcopenia. Old adults with sarcopenia more likely have higher body fat percentage, lower BMI and lower cognitive function compared with those without this condition.
Collapse
Affiliation(s)
- Jiazhi Wang
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Changge Liu
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Lin Zhang
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Ning Liu
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Lei Wang
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Jingqiong Wu
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Yizhao Wang
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Huimin Hao
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Longjun Cao
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China
| | - Shilei Yuan
- Tianjin Yanan Hospital, No. 45 Yashi Road, Nankai District, Tianjin, P.R. China
| | - Liping Huang
- Tianjin University of Sport, No.16 Donghai Road, West Tuanbo New Town, Jinghai District, Tianjin, P.R. China.
| |
Collapse
|
33
|
Kim BJ. Effects of Muscles on Bone Metabolism—with a Focus on Myokines. Ann Geriatr Med Res 2022; 26:63-71. [PMID: 35722780 PMCID: PMC9271391 DOI: 10.4235/agmr.22.0054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/07/2022] [Indexed: 11/10/2022] Open
Abstract
Skeletal muscles and bones, the largest tissues in the body of a non-obese person, comprise the musculoskeletal system, which allows mobility and protects internal organs. Although muscles and bones are closely related throughout life, observations during development and aging and in human and animal disuse models have revealed the synchronization of tissue mass such that muscle phenotype changes precede alterations in bone mineral density and strength. This review discussed that mechanical forces, which have been the traditional research focus, are not the only mechanism by which muscle-derived signals may affect bone metabolism and emphasized the significance of skeletal muscles as an endocrine organ that secretes bone-regulatory factors. Consequently, both mechanical and biochemical aspects should be considered to fully understand muscle–bone crosstalk. This review also suggested that specific myokines could be ideal therapeutic targets for osteoporosis to both increase bone formation and reduce bone resorption; moreover, these myokines could also be potential circulating biomarkers to predict musculoskeletal health.
Collapse
Affiliation(s)
- Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Corresponding Authors: Beom-Jun Kim, MD, PhD Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea E-mail:
| |
Collapse
|
34
|
Therapeutic Benefit in Rheumatoid Cachexia Illustrated Using a Novel Primary Human Triple Cell Coculture Model. Int J Inflam 2022; 2022:1524913. [PMID: 35693848 PMCID: PMC9184217 DOI: 10.1155/2022/1524913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 11/21/2022] Open
Abstract
Background The loss of muscle mass in rheumatoid arthritis (RA), termed rheumatoid cachexia, is predicted to result from the complex interactions between different cell types involved in the maintenance of skeletal muscle mass, namely, myoblasts, fibroblasts, and macrophages. The complexity within the muscle is further highlighted by the incidence of nonresponsiveness to current RA treatment strategies. Method This study aimed at determining differences in the cellular responses in a novel human primary cell triple coculture model exposed to serum collected from nonarthritic controls (NC), RA treatment naïve (RATN), and RA treatment-nonresponding (RATNR) patients. Bone morphogenetic protein-7 (BMP-7) was investigated as a treatment option. Results Plasma analysis indicated that samples were indeed representative of healthy and RA patients—notably, the RATNR patients additionally exhibited dysregulated IL-6/IL-10 correlations. Coculture exposure to serum from RATNR patients demonstrated increased cellular growth (p < 0.001), while both hepatocyte growth factor (p < 0.01) and follistatin (p < 0.001) were reduced when compared to NC. Furthermore, decreased concentration of markers of extracellular matrix formation, transforming growth factor-β (TGF-β; p < 0.05) and fibronectin (p < 0.001), but increased collagen IV (p < 0.01) was observed following RATNR serum exposure. Under healthy conditions, BMP-7 exhibited potentially beneficial results in reducing fibrosis-generating TGF-β (p < 0.05) and fibronectin (p < 0.05). BMP-7 further exhibited protective potential in the RA groups through reversing the aberrant tendencies observed especially in the RATNR serum-exposed group. Conclusion Exposure of the triple coculture to RATN and RATNR serum resulted in dysregulated myoblast proliferation and growth, and ECM impairment, which was reversed by BMP-7 treatment.
Collapse
|
35
|
Li G, Jin B, Fan Z. Mechanisms Involved in Gut Microbiota Regulation of Skeletal Muscle. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2151191. [PMID: 35633886 PMCID: PMC9132697 DOI: 10.1155/2022/2151191] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
Abstract
Skeletal muscle is one of the largest organs in the body and is essential for maintaining quality of life. Loss of skeletal muscle mass and function can lead to a range of adverse consequences. The gut microbiota can interact with skeletal muscle by regulating a variety of processes that affect host physiology, including inflammatory immunity, protein anabolism, energy, lipids, neuromuscular connectivity, oxidative stress, mitochondrial function, and endocrine and insulin resistance. It is proposed that the gut microbiota plays a role in the direction of skeletal muscle mass and work. Even though the notion of the gut microbiota-muscle axis (gut-muscle axis) has been postulated, its causal link is still unknown. The impact of the gut microbiota on skeletal muscle function and quality is described in detail in this review.
Collapse
Affiliation(s)
- Guangyao Li
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Binghui Jin
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Zhe Fan
- Department of General Surgery, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People's Hospital of Dalian, Dalian Medical University, Dalian, China
| |
Collapse
|
36
|
Sabouri M, Taghibeikzadehbadr P, Shabkhiz F, Izanloo Z, Shaghaghi FA. Effect of eccentric and concentric contraction mode on myogenic regulatory factors expression in human vastus lateralis muscle. J Muscle Res Cell Motil 2022; 43:9-20. [PMID: 35018575 DOI: 10.1007/s10974-021-09613-x] [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: 07/05/2021] [Accepted: 12/01/2021] [Indexed: 11/26/2022]
Abstract
Skeletal muscle contractions are caused to release myokines by muscle fiber. This study investigated the myogenic regulatory factors, as MHC I, IIA, IIX, Myo-D, MRF4, Murf, Atrogin-1, Decorin, Myonection, and IL-15 mRNA expression in the response of eccentric vs concentric contraction. Eighteen healthy men were randomly divided into two eccentric and concentric groups, each of 9 persons. Isokinetic contraction protocols included maximal single-leg eccentric or concentric knee extension tasks at 60°/s with the dominant leg. Contractions consisted of a maximum of 12 sets of 10 reps, and the rest time between each set was 30 s. The baseline biopsy was performed 4 weeks before the study, and post-test biopsies were taken immediately after exercise protocols from the vastus lateralis muscle. The gene expression levels were evaluated using Real-Time PCR methods. The eccentric group showed a significantly lower RPE score than the concentric group (P ≤ 0.05). A significant difference in MyoD, MRF4, Myonection, and Decorin mRNA, were observed following eccentric or concentric contractions (P ≤ 0.05). The MHC I, MHC IIA, IL-15 mRNA has been changed significantly compared to the pre-exercise in the concentric group (P ≤ 0.05). While only MHC IIX and Atrogin-1 mRNA changed significantly in the eccentric group (P ≤ 0.05). Additionally, the results showed a significant difference in MyoD, MRF4, IL-15, and Decorin at the follow-up values between eccentric or concentric groups (P ≤ 0.05). Our findings highlight the growing importance of elucidating the different responses of muscle growth factors associated with a myogenic activity such as MHC IIA, Decorin, IL-15, Myonectin, Decorin, MuRF1, and MHC IIX mRNA in following various types of exercise.
Collapse
Affiliation(s)
- Mostafa Sabouri
- Department of Exercise Physiology & Health Science, University of Tehran, Tehran, Iran.
| | | | - Fatemeh Shabkhiz
- Department of Exercise Physiology & Health Science, University of Tehran, Tehran, Iran
| | - Zahra Izanloo
- Department of Sport Science, Faculty of Human Science, University of Bojnord, Bojnord, Iran
| | | |
Collapse
|
37
|
Yates AG, Pink RC, Erdbrügger U, Siljander PR, Dellar ER, Pantazi P, Akbar N, Cooke WR, Vatish M, Dias‐Neto E, Anthony DC, Couch Y. In sickness and in health: The functional role of extracellular vesicles in physiology and pathology in vivo: Part I: Health and Normal Physiology: Part I: Health and Normal Physiology. J Extracell Vesicles 2022; 11:e12151. [PMID: 35041249 PMCID: PMC8765331 DOI: 10.1002/jev2.12151] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
Previously thought to be nothing more than cellular debris, extracellular vesicles (EVs) are now known to mediate physiological and pathological functions throughout the body. We now understand more about their capacity to transfer nucleic acids and proteins between distant organs, the interaction of their surface proteins with target cells, and the role of vesicle-bound lipids in health and disease. To date, most observations have been made in reductionist cell culture systems, or as snapshots from patient cohorts. The heterogenous population of vesicles produced in vivo likely act in concert to mediate both beneficial and detrimental effects. EVs play crucial roles in both the pathogenesis of diseases, from cancer to neurodegenerative disease, as well as in the maintenance of system and organ homeostasis. This two-part review draws on the expertise of researchers working in the field of EV biology and aims to cover the functional role of EVs in physiology and pathology. Part I will outline the role of EVs in normal physiology.
Collapse
Affiliation(s)
- Abi G. Yates
- Department of PharmacologyUniversity of OxfordOxfordUK
- School of Biomedical SciencesFaculty of MedicineUniversity of QueenslandSt LuciaAustralia
| | - Ryan C. Pink
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Uta Erdbrügger
- Department of Medicine, Division of NephrologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Pia R‐M. Siljander
- Molecular and Integrative Biosciences Research ProgrammeFaculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
| | - Elizabeth R. Dellar
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Paschalia Pantazi
- Department of Biological and Medical SciencesFaculty of Health and Life SciencesOxford Brookes UniversityHeadington CampusOxfordUK
| | - Naveed Akbar
- Division of Cardiovascular Medicine, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - William R. Cooke
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Manu Vatish
- Nuffield Department of Women's and Reproductive HealthUniversity of OxfordOxfordUK
| | - Emmanuel Dias‐Neto
- Laboratory of Medical Genomics. A.C. Camargo Cancer CentreSão PauloBrazil
- Laboratory of Neurosciences (LIM‐27) Institute of PsychiatrySão Paulo Medical SchoolSão PauloBrazil
| | | | - Yvonne Couch
- Acute Stroke Programme ‐ Radcliffe Department of MedicineUniversity of OxfordJohn Radcliffe Hospital, HeadingtonOxfordUK
| |
Collapse
|
38
|
Secretome from In Vitro Mechanically Loaded Myoblasts Induces Tenocyte Migration, Transition to a Fibroblastic Phenotype and Suppression of Collagen Production. Int J Mol Sci 2021; 22:ijms222313089. [PMID: 34884895 PMCID: PMC8657858 DOI: 10.3390/ijms222313089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
It is known that mechanical loading of muscles increases the strength of healing tendon tissue, but the mechanism involved remains elusive. We hypothesized that the secretome from myoblasts in co-culture with tenocytes affects tenocyte migration, cell phenotype, and collagen (Col) production and that the effect is dependent on different types of mechanical loading of myoblasts. To test this, we used an in vitro indirect transwell co-culture system. Myoblasts were mechanically loaded using the FlexCell® Tension system. Tenocyte cell migration, proliferation, apoptosis, collagen production, and several tenocyte markers were measured. The secretome from myoblasts decreased the Col I/III ratio and increased the expression of tenocyte specific markers as compared with tenocytes cultured alone. The secretome from statically loaded myoblasts significantly enhanced tenocyte migration and Col I/III ratio as compared with dynamic loading and controls. In addition, the secretome from statically loaded myoblasts induced tenocytes towards a myofibroblast-like phenotype. Taken together, these results demonstrate that the secretome from statically loaded myoblasts has a profound influence on tenocytes, affecting parameters that are related to the tendon healing process.
Collapse
|
39
|
Boido M, De Amicis E, Mareschi K, Fagioli F, Vercelli A. Organotypic spinal cord cultures: An <em>in vitro</em> 3D model to preliminary screen treatments for spinal muscular atrophy. Eur J Histochem 2021; 65. [PMID: 34734684 PMCID: PMC8586821 DOI: 10.4081/ejh.2021.3294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/24/2021] [Indexed: 11/23/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a severe neuromuscular disease affecting children, due to mutation/deletion of survival motor neuron 1 (SMN1) gene. The lack of functional protein SMN determines motor neuron (MN) degeneration and skeletal muscle atrophy, leading to premature death due to respiratory failure. Nowadays, the Food and Drug Administration approved the administration of three drugs, aiming at increasing the SMN production: although assuring noteworthy results, all these therapies show some non-negligible limitations, making essential the identification of alternative/synergistic therapeutic strategies. To offer a valuable in vitro experimental model for easily performing preliminary screenings of alternative promising treatments, we optimized an organotypic spinal cord culture (derived from murine spinal cord slices), which well recapitulates the pathogenetic features of SMA. Then, to validate the model, we tested the effects of human mesenchymal stem cells (hMSCs) or murine C2C12 cells (a mouse skeletal myoblast cell line) conditioned media: 1/3 of conditioned medium (obtained from either hMSCs or C2C12 cells) was added to the conventional medium of the organotypic culture and maintained for 7 days. Then the slices were fixed and immunoreacted to evaluate the MN survival. In particular we observed that the C2C12 and hMSCs conditioned media positively influenced the MN soma size and the axonal length respectively, without modulating the glial activation. These data suggest that trophic factors released by MSCs or muscular cells can exert beneficial effects, by acting on different targets, and confirm the reliability of the model. Overall, we propose the organotypic spinal cord culture as an excellent tool to preliminarily screen molecules and drugs before moving to in vivo models, in this way partly reducing the use of animals and the costs.
Collapse
Affiliation(s)
- Marina Boido
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience "Rita Levi Montalcini", University of Turin.
| | - Elena De Amicis
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience "Rita Levi Montalcini", University of Turin.
| | - Katia Mareschi
- Department of Public Health and Paediatrics, University of Turin; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin.
| | - Franca Fagioli
- Department of Public Health and Paediatrics, University of Turin; Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Division, Regina Margherita Children's Hospital, City of Health and Science of Turin.
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi, Department of Neuroscience "Rita Levi Montalcini", University of Turin.
| |
Collapse
|
40
|
Paeschke S, Winter K, Bechmann I, Klöting N, Blüher M, Baum P, Kosacka J, Nowicki M. Leptin Receptor-Deficient db/db Mice Show Significant Heterogeneity in Response to High Non-heme Iron Diet. Front Nutr 2021; 8:741249. [PMID: 34646852 PMCID: PMC8503537 DOI: 10.3389/fnut.2021.741249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022] Open
Abstract
Recent studies have shown an association between iron homeostasis, obesity and diabetes. In this work, we investigated the differences in the metabolic status and inflammation in liver, pancreas and visceral adipose tissue of leptin receptor-deficient db/db mice dependent on high iron concentration diet. 3-month-old male BKS-Leprdb/db/JOrlRj (db/db) mice were divided into two groups, which were fed with different diets containing high iron (29 g/kg, n = 57) or standard iron (0.178 g/kg; n = 42) concentrations for 4 months. As anticipated, standard iron-fed db/db mice developed obesity and diabetes. However, high iron-fed mice exhibited a wide heterogeneity. By dividing into two subgroups at the diabetes level, non-diabetic subgroup 1 (<13.5 mmol/l, n = 30) significantly differed from diabetic subgroup two (>13.5 mmol/l, n = 27). Blood glucose concentration, HbA1c value, inflammation markers interleukin six and tumor necrosis factor α and heme oxygenase one in visceral adipose tissue were reduced in subgroup one compared to subgroup two. In contrast, body weight, C-peptide, serum insulin and serum iron concentrations, pancreatic islet and signal ratio as well as cholesterol, LDL and HDL levels were enhanced in subgroup one. While these significant differences require further studies and explanation, our results might also explain the often-contradictory results of the metabolic studies with db/db mice.
Collapse
Affiliation(s)
- Sabine Paeschke
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum Munchen at the University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum Munchen at the University of Leipzig, Leipzig, Germany.,Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Petra Baum
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Joanna Kosacka
- Department of Medicine, University of Leipzig, Leipzig, Germany.,Applied Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Marcin Nowicki
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
41
|
Howard AC, Mir D, Snow S, Horrocks J, Sayed H, Ma Z, Rogers AN. Anabolic Function Downstream of TOR Controls Trade-offs Between Longevity and Reproduction at the Level of Specific Tissues in C. elegans. FRONTIERS IN AGING 2021; 2:725068. [PMID: 35340273 PMCID: PMC8953723 DOI: 10.3389/fragi.2021.725068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/27/2021] [Indexed: 11/20/2022]
Abstract
As the most energetically expensive cellular process, translation must be finely tuned to environmental conditions. Dietary restriction attenuates signaling through the nutrient sensing mTOR pathway, which reduces translation and redirects resources to preserve the soma. These responses are associated with increased lifespan but also anabolic impairment, phenotypes also observed when translation is genetically suppressed. Here, we restricted translation downstream of mTOR separately in major tissues in C. elegans to better understand their roles in systemic adaptation and whether consequences to anabolic impairment were separable from positive effects on lifespan. Lowering translation in neurons, hypodermis, or germline tissue led to increased lifespan under well-fed conditions and improved survival upon withdrawal of food, indicating that these are key tissues coordinating enhanced survival when protein synthesis is reduced. Surprisingly, lowering translation in body muscle during development shortened lifespan while accelerating and increasing reproduction, a reversal of phenotypic trade-offs associated with systemic translation suppression. Suppressing mTORC1 selectively in body muscle also increased reproduction while slowing motility during development. In nature, this may be indicative of reduced energy expenditure related to foraging, acting as a "GO!" signal for reproduction. Together, results indicate that low translation in different tissues helps direct distinct systemic adaptations and suggest that unknown endocrine signals mediate these responses. Furthermore, mTOR or translation inhibitory therapeutics that target specific tissues may achieve desired interventions to aging without loss of whole-body anabolism.
Collapse
Affiliation(s)
- Amber C. Howard
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
- Department of Natural Sciences, Middle Georgia State University, Cochran, GA, United States
| | - Dilawar Mir
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Santina Snow
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
- Department of Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, United States
| | - Jordan Horrocks
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Hussein Sayed
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Zhengxin Ma
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| | - Aric N. Rogers
- Mount Desert Island Biological Laboratory, Davis Center for Regenerative Biology and Medicine, Bar Harbor, ME, United States
| |
Collapse
|
42
|
Mancinelli R, Checcaglini F, Coscia F, Gigliotti P, Fulle S, Fanò-Illic G. Biological Aspects of Selected Myokines in Skeletal Muscle: Focus on Aging. Int J Mol Sci 2021; 22:8520. [PMID: 34445222 PMCID: PMC8395159 DOI: 10.3390/ijms22168520] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
In the last decade, clear evidence has emerged that the cellular components of skeletal muscle are important sites for the release of proteins and peptides called "myokines", suggesting that skeletal muscle plays the role of a secretory organ. After their secretion by muscles, these factors serve many biological functions, including the exertion of complex autocrine, paracrine and/or endocrine effects. In sum, myokines affect complex multi-organ processes, such as skeletal muscle trophism, metabolism, angiogenesis and immunological response to different physiological (physical activity, aging, etc.) or pathological states (cachexia, dysmetabolic conditions, chronic inflammation, etc.). The aim of this review is to describe in detail a number of myokines that are, to varying degrees, involved in skeletal muscle aging processes and belong to the group of proteins present in the functional environment surrounding the muscle cell known as the "Niche". The particular myokines described are those that, acting both from within the cell and in an autocrine manner, have a defined relationship with the modulation of oxidative stress in muscle cells (mature or stem) involved in the regulatory (metabolic or regenerative) processes of muscle aging. Myostatin, IGF-1, NGF, S100 and irisin are examples of specific myokines that have peculiar features in their mechanisms of action. In particular, the potential role of one of the most recently characterized myokines-irisin, directly linked to an active lifestyle-in reducing if not reversing senescence-induced oxidative damage is discussed in terms of its possible application as an agent able to counteract the deleterious effects of muscle aging.
Collapse
Affiliation(s)
- Rosa Mancinelli
- Department of Neuroscience Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (R.M.); (S.F.)
- IIM-Interuniversity Institute of Myology, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Franco Checcaglini
- Free University of Alcatraz, Santa Cristina di Gubbio, 06100 Perugia, Italy;
| | - Francesco Coscia
- Department of Medicine, Laboratory of Sport Physiology, University of Perugia, 39038 San Candido-Innichen, Italy; (F.C.); (P.G.)
| | - Paola Gigliotti
- Department of Medicine, Laboratory of Sport Physiology, University of Perugia, 39038 San Candido-Innichen, Italy; (F.C.); (P.G.)
| | - Stefania Fulle
- Department of Neuroscience Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (R.M.); (S.F.)
- IIM-Interuniversity Institute of Myology, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgio Fanò-Illic
- Department of Neuroscience Imaging and Clinical Sciences, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (R.M.); (S.F.)
- IIM-Interuniversity Institute of Myology, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
- Free University of Alcatraz, Santa Cristina di Gubbio, 06100 Perugia, Italy;
- A&C M-C Foundation for Translational Myology, 35100 Padova, Italy
| |
Collapse
|
43
|
Gonzalez-Franquesa A, Peijs L, Cervone DT, Koçana C, Zierath JR, Deshmukh AS. Insulin and 5-Aminoimidazole-4-Carboxamide Ribonucleotide (AICAR) Differentially Regulate the Skeletal Muscle Cell Secretome. Proteomes 2021; 9:37. [PMID: 34449730 PMCID: PMC8396280 DOI: 10.3390/proteomes9030037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle is a major contributor to whole-body glucose homeostasis and is an important endocrine organ. To date, few studies have undertaken the large-scale identification of skeletal muscle-derived secreted proteins (myokines), particularly in response to stimuli that activate pathways governing energy metabolism in health and disease. Whereas the AMP-activated protein kinase (AMPK) and insulin-signaling pathways have received notable attention for their ability to independently regulate skeletal muscle substrate metabolism, little work has examined their ability to re-pattern the secretome. The present study coupled the use of high-resolution MS-based proteomics and bioinformatics analysis of conditioned media derived from 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR-an AMPK activator)- and insulin-treated differentiated C2C12 myotubes. We quantified 858 secreted proteins, including cytokines and growth factors such as fibroblast growth factor-21 (Fgf21). We identified 377 and 118 proteins that were significantly altered by insulin and AICAR treatment, respectively. Notably, the family of insulin growth factor binding-proteins (Igfbp) was differentially regulated by each treatment. Insulin- but not AICAR-induced conditioned media increased the mitochondrial respiratory capacity of myotubes, potentially via secreted factors. These findings may serve as an important resource to elucidate secondary metabolic effects of insulin and AICAR stimulation in skeletal muscle.
Collapse
Affiliation(s)
- Alba Gonzalez-Franquesa
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (A.G.-F.); (L.P.); (D.T.C.); (C.K.); (J.R.Z.)
| | - Lone Peijs
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (A.G.-F.); (L.P.); (D.T.C.); (C.K.); (J.R.Z.)
| | - Daniel T. Cervone
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (A.G.-F.); (L.P.); (D.T.C.); (C.K.); (J.R.Z.)
| | - Ceren Koçana
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (A.G.-F.); (L.P.); (D.T.C.); (C.K.); (J.R.Z.)
| | - Juleen R. Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (A.G.-F.); (L.P.); (D.T.C.); (C.K.); (J.R.Z.)
- Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Atul S. Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; (A.G.-F.); (L.P.); (D.T.C.); (C.K.); (J.R.Z.)
- Clinical Proteomics, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
44
|
Debaud C, Tseng HW, Chedik M, Kulina I, Genêt F, Ruitenberg MJ, Levesque JP. Local and Systemic Factors Drive Ectopic Osteogenesis in Regenerating Muscles of Spinal-Cord-Injured Mice in a Lesion-Level-Dependent Manner. J Neurotrauma 2021; 38:2162-2175. [PMID: 33913747 DOI: 10.1089/neu.2021.0058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neuroimmune dysfunction is thought to promote the development of several acute and chronic complications in spinal cord injury (SCI) patients. Putative roles for adrenal stress hormones and catecholamines are increasingly being recognized, yet how these adversely affect peripheral tissue homeostasis and repair under SCI conditions remains elusive. Here, we investigated their influence in a mouse model of SCI with acquired neurogenic heterotopic ossification. We show that spinal cord lesions differentially influence muscular regeneration in a level-dependent manner and through a complex multi-step process that creates an osteopermissive environment within the first hours of injury. This cascade of events is shown to critically involve adrenergic signals and drive the acute release of the neuropeptide, substance P. Our findings generate new insights into the kinetics and processes that govern SCI-induced deregulations in skeletal muscle homeostasis and regeneration, thereby aiding the development of sequential therapeutic strategies that can prevent or attenuate neuromusculoskeletal complications in SCI patients.
Collapse
Affiliation(s)
- Charlotte Debaud
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
- Spine Division, Orthopaedic Surgery Department, Queensland Health, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
- Université de Versailles Saint Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé-Simone Veil, Montigny-le-Bretonneux, France
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Hsu-Wen Tseng
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Malha Chedik
- Université de Versailles Saint Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé-Simone Veil, Montigny-le-Bretonneux, France
| | - Irina Kulina
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - François Genêt
- Université de Versailles Saint Quentin en Yvelines, U1179 INSERM, UFR des Sciences de la Santé-Simone Veil, Montigny-le-Bretonneux, France
- Service de Réhabilitation, Hôpital Raymond Poincaré, APHP, CIC-IT 1429, Garches, France
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Saint Lucia, Queensland, Australia
| | - Jean-Pierre Levesque
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| |
Collapse
|
45
|
Reiss J, Robertson S, Suzuki M. Cell Sources for Cultivated Meat: Applications and Considerations throughout the Production Workflow. Int J Mol Sci 2021; 22:7513. [PMID: 34299132 PMCID: PMC8307620 DOI: 10.3390/ijms22147513] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Cellular agriculture is an emerging scientific discipline that leverages the existing principles behind stem cell biology, tissue engineering, and animal sciences to create agricultural products from cells in vitro. Cultivated meat, also known as clean meat or cultured meat, is a prominent subfield of cellular agriculture that possesses promising potential to alleviate the negative externalities associated with conventional meat production by producing meat in vitro instead of from slaughter. A core consideration when producing cultivated meat is cell sourcing. Specifically, developing livestock cell sources that possess the necessary proliferative capacity and differentiation potential for cultivated meat production is a key technical component that must be optimized to enable scale-up for commercial production of cultivated meat. There are several possible approaches to develop cell sources for cultivated meat production, each possessing certain advantages and disadvantages. This review will discuss the current cell sources used for cultivated meat production and remaining challenges that need to be overcome to achieve scale-up of cultivated meat for commercial production. We will also discuss cell-focused considerations in other components of the cultivated meat production workflow, namely, culture medium composition, bioreactor expansion, and biomaterial tissue scaffolding.
Collapse
Affiliation(s)
- Jacob Reiss
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (J.R.); (S.R.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
46
|
Gagaoua M, Warner RD, Purslow P, Ramanathan R, Mullen AM, López-Pedrouso M, Franco D, Lorenzo JM, Tomasevic I, Picard B, Troy D, Terlouw EMC. Dark-cutting beef: A brief review and an integromics meta-analysis at the proteome level to decipher the underlying pathways. Meat Sci 2021; 181:108611. [PMID: 34157500 DOI: 10.1016/j.meatsci.2021.108611] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 01/06/2023]
Abstract
Comprehensive characterization of the post-mortem muscle proteome defines a fundamental goal in meat proteomics. During the last decade, proteomics tools have been applied in the field of foodomics to help decipher factors underpinning meat quality variations and to enlighten us, through data-driven methods, on the underlying mechanisms leading to meat quality defects such as dark-cutting meat known also as dark, firm and dry (DFD) meat. In cattle, several proteomics studies have focused on the extent to which changes in the post-mortem muscle proteome relate to dark-cutting beef development. The present data-mining study firstly reviews proteomics studies which investigated dark-cutting beef, and secondly, gathers the protein biomarkers that differ between dark-cutting versus beef with normal-pH in a unique repertoire. A list of 130 proteins from eight eligible studies was curated and mined through bioinformatics for Gene Ontology annotations, molecular pathways enrichments, secretome analysis and biological pathways comparisons to normal beef color from a previous meta-analysis. The major biological pathways underpinning dark-cutting beef at the proteome level have been described and deeply discussed in this integromics study.
Collapse
Affiliation(s)
- Mohammed Gagaoua
- Food Quality and Sensory Science Department, Teagasc Food Research Centre, Ashtown, Dublin 15, Ireland.
| | - Robyn D Warner
- School of Agriculture and Food, Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Peter Purslow
- Centro de Investigacion Veterinaria de Tandil (CIVETAN), Universidad Nacional del Centro de la Provincia de Buenos Aires, Tandil B7001BBO, Argentina
| | - Ranjith Ramanathan
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Anne Maria Mullen
- Food Quality and Sensory Science Department, Teagasc Food Research Centre, Ashtown, Dublin 15, Ireland
| | - Maria López-Pedrouso
- Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, 15872 Santiago de Compostela, Spain
| | - Daniel Franco
- Centro Tecnológico de la Carne de Galicia, rúa Galicia n° 4, Parque Tecnológico de Galicia, San Cibrao das Viñas 32900, Ourense, Spain
| | - José M Lorenzo
- Centro Tecnológico de la Carne de Galicia, rúa Galicia n° 4, Parque Tecnológico de Galicia, San Cibrao das Viñas 32900, Ourense, Spain; Área de Tecnología de los Alimentos, Facultad de Ciencias de Ourense, Universidad de Vigo, 32004 Ourense, Spain
| | - Igor Tomasevic
- University of Belgrade, Faculty of Agriculture, Nemanjina 6, 11080, Belgrade, Serbia
| | - Brigitte Picard
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| | - Declan Troy
- Food Quality and Sensory Science Department, Teagasc Food Research Centre, Ashtown, Dublin 15, Ireland
| | - E M Claudia Terlouw
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMR Herbivores, F-63122 Saint-Genès-Champanelle, France
| |
Collapse
|
47
|
Lee B, Shin M, Park Y, Won SY, Cho KS. Physical Exercise-Induced Myokines in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22115795. [PMID: 34071457 PMCID: PMC8198301 DOI: 10.3390/ijms22115795] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative diseases (NDs), such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS), are disorders characterized by progressive degeneration of the nervous system. Currently, there is no disease-modifying treatments for most NDs. Meanwhile, numerous studies conducted on human and animal models over the past decades have showed that exercises had beneficial effects on NDs. Inter-tissue communication by myokine, a peptide produced and secreted by skeletal muscles during exercise, is thought to be an important underlying mechanism for the advantages. Here, we reviewed studies about the effects of myokines regulated by exercise on NDs and their mechanisms. Myokines could exert beneficial effects on NDs through a variety of regulatory mechanisms, including cell survival, neurogenesis, neuroinflammation, proteostasis, oxidative stress, and protein modification. Studies on exercise-induced myokines are expected to provide a novel strategy for treating NDs, for which there are no adequate treatments nowadays. To date, only a few myokines have been investigated for their effects on NDs and studies on mechanisms involved in them are in their infancy. Therefore, future studies are needed to discover more myokines and test their effects on NDs.
Collapse
Affiliation(s)
- Banseok Lee
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - Myeongcheol Shin
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - Youngjae Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
| | - So-Yoon Won
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
- Korea Hemp Institute, Konkuk University, Seoul 05029, Korea
- Correspondence: (S.-Y.W.); (K.S.C.); Tel.: +82-10-3688-5474 (S.-Y.W.); Tel.: +82-2-450-3424 (K.S.C.)
| | - Kyoung Sang Cho
- Department of Biological Sciences, Konkuk University, Seoul 05029, Korea; (B.L.); (M.S.); (Y.P.)
- Korea Hemp Institute, Konkuk University, Seoul 05029, Korea
- Correspondence: (S.-Y.W.); (K.S.C.); Tel.: +82-10-3688-5474 (S.-Y.W.); Tel.: +82-2-450-3424 (K.S.C.)
| |
Collapse
|
48
|
Marques-Aleixo I, Beleza J, Sampaio A, Stevanović J, Coxito P, Gonçalves I, Ascensão A, Magalhães J. Preventive and Therapeutic Potential of Physical Exercise in Neurodegenerative Diseases. Antioxid Redox Signal 2021; 34:674-693. [PMID: 32159378 DOI: 10.1089/ars.2020.8075] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: The prevalence and incidence of age-related neurodegenerative diseases (NDDs) tend to increase along with the enhanced average of the world life expectancy. NDDs are a major cause of morbidity and disability, affecting the health care, social and economic systems with a significant impact. Critical Issues and Recent Advances: Despite the worldwide burden of NDDs and the ongoing research efforts to increase the underlying molecular mechanisms involved in NDD pathophysiologies, pharmacological therapies have been presenting merely narrow benefits. On the contrary, absent of detrimental side effects but growing merits, regular physical exercise (PE) has been considered a prone pleiotropic nonpharmacological alternative able to modulate brain structure and function, thereby stimulating a healthier and "fitness" neurological phenotype. Future Directions: This review summarizes the state of the art of some peripheral and central-related mechanisms that underlie the impact of PE on brain plasticity as well as its relevance for the prevention and/or treatment of NDDs. Nevertheless, further studies are needed to better clarify the molecular signaling pathways associated with muscle contractions-related myokines release and its plausible positive effects in the brain. In addition, particular focus of research should address the role of PE in the modulation of mitochondrial metabolism and oxidative stress in the context of NDDs.
Collapse
Affiliation(s)
- Inês Marques-Aleixo
- Faculty of Psychology, Education and Sports, Lusofona University of Porto, Porto, Portugal.,Laboratory of Metabolism and Exercise (LaMetEx), Research Center in Physical Activity Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADEUP), Porto, Portugal
| | - Jorge Beleza
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Arnaldina Sampaio
- Laboratory of Metabolism and Exercise (LaMetEx), Research Center in Physical Activity Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADEUP), Porto, Portugal
| | - Jelena Stevanović
- Laboratory of Metabolism and Exercise (LaMetEx), Research Center in Physical Activity Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADEUP), Porto, Portugal
| | - Pedro Coxito
- Laboratory of Metabolism and Exercise (LaMetEx), Research Center in Physical Activity Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADEUP), Porto, Portugal
| | | | - António Ascensão
- Laboratory of Metabolism and Exercise (LaMetEx), Research Center in Physical Activity Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADEUP), Porto, Portugal
| | - José Magalhães
- Laboratory of Metabolism and Exercise (LaMetEx), Research Center in Physical Activity Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADEUP), Porto, Portugal
| |
Collapse
|
49
|
Ratnayake D, Nguyen PD, Rossello FJ, Wimmer VC, Tan JL, Galvis LA, Julier Z, Wood AJ, Boudier T, Isiaku AI, Berger S, Oorschot V, Sonntag C, Rogers KL, Marcelle C, Lieschke GJ, Martino MM, Bakkers J, Currie PD. Macrophages provide a transient muscle stem cell niche via NAMPT secretion. Nature 2021; 591:281-287. [PMID: 33568815 DOI: 10.1038/s41586-021-03199-7] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/07/2021] [Indexed: 01/30/2023]
Abstract
Skeletal muscle regenerates through the activation of resident stem cells. Termed satellite cells, these normally quiescent cells are induced to proliferate by wound-derived signals1. Identifying the source and nature of these cues has been hampered by an inability to visualize the complex cell interactions that occur within the wound. Here we use muscle injury models in zebrafish to systematically capture the interactions between satellite cells and the innate immune system after injury, in real time, throughout the repair process. This analysis revealed that a specific subset of macrophages 'dwell' within the injury, establishing a transient but obligate niche for stem cell proliferation. Single-cell profiling identified proliferative signals that are secreted by dwelling macrophages, which include the cytokine nicotinamide phosphoribosyltransferase (Nampt, which is also known as visfatin or PBEF in humans). Nampt secretion from the macrophage niche is required for muscle regeneration, acting through the C-C motif chemokine receptor type 5 (Ccr5), which is expressed on muscle stem cells. This analysis shows that in addition to their ability to modulate the immune response, specific macrophage populations also provide a transient stem-cell-activating niche, directly supplying proliferation-inducing cues that govern the repair process that is mediated by muscle stem cells. This study demonstrates that macrophage-derived niche signals for muscle stem cells, such as NAMPT, can be applied as new therapeutic modalities for skeletal muscle injury and disease.
Collapse
Affiliation(s)
- Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Phong D Nguyen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Fernando J Rossello
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,University of Melbourne Centre for Cancer Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Verena C Wimmer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jean L Tan
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Laura A Galvis
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Institut NeuroMyoGène (INMG), University Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Ziad Julier
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Thomas Boudier
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdulsalam I Isiaku
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Silke Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Viola Oorschot
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Victoria, Australia.,European Molecular Biology Laboratory, Electron Microscopy Core Facility, Heidelberg, Germany
| | - Carmen Sonntag
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Christophe Marcelle
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Institut NeuroMyoGène (INMG), University Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Mikaël M Martino
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Jeroen Bakkers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. .,EMBL Australia, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
50
|
Sarcopenia and Cognitive Function: Role of Myokines in Muscle Brain Cross-Talk. Life (Basel) 2021; 11:life11020173. [PMID: 33672427 PMCID: PMC7926334 DOI: 10.3390/life11020173] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 02/07/2023] Open
Abstract
Sarcopenia is a geriatric syndrome characterized by the progressive degeneration of muscle mass and function, and it is associated with severe complications, which are falls, functional decline, frailty, and mortality. Sarcopenia is associated with cognitive impairment, defined as a decline in one or more cognitive domains as language, memory, reasoning, social cognition, planning, making decisions, and solving problems. Although the exact mechanism relating to sarcopenia and cognitive function has not yet been defined, several studies have shown that skeletal muscle produces and secrete molecules, called myokines, that regulate brain functions, including mood, learning, locomotor activity, and neuronal injury protection, showing the existence of muscle-brain cross-talk. Moreover, studies conducted on physical exercise supported the existence of muscle-brain cross-talk, showing how physical activity, changing myokines' circulating levels, exerts beneficial effects on the brain. The review mainly focuses on describing the role of myokines on brain function and their involvement in cognitive impairment in sarcopenia.
Collapse
|