1
|
Meacci E, Chirco A, Garcia-Gil M. Potential Vitamin E Signaling Mediators in Skeletal Muscle. Antioxidants (Basel) 2024; 13:1383. [PMID: 39594525 PMCID: PMC11591548 DOI: 10.3390/antiox13111383] [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: 09/14/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Vitamin E (Vit E) deficiency studies underline the relevance of this vitamin in skeletal muscle (SkM) homeostasis. The knowledge of the effectors and modulators of Vit E action in SkM cells is limited, especially in aging and chronic diseases characterized by a decline in musculoskeletal health. Vit E comprises eight fat-soluble compounds grouped into tocopherols and tocotrienols, which share the basic chemical structure but show different biological properties and potentials to prevent diseases. Vit E has antioxidant and non-antioxidant activities and both favorable and adverse effects depending on the specific conditions and tissues. In this review, we focus on the actual knowledge of Vit E forms in SkM functions and new potential signaling effectors (i.e., bioactive sphingolipids and myokines). The possible advantages of Vit E supplementation in counteracting SkM dysfunctions in sarcopenia and under microgravity will also be discussed.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Department of Experimental and clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Firenze, Italy
- Interuniversity Institute of Myology, University of Florence, 50134 Firenze, Italy
| | - Antony Chirco
- Department of Experimental and clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Firenze, Italy
| | - Mercedes Garcia-Gil
- Department of Biology, Unit of Physiology, University of Pisa, Via S. Zeno 31, 56127 Pisa, Italy;
| |
Collapse
|
2
|
Zhao D, Zhuang W, Wang Y, Xu X, Qiao L. In-depth mass spectrometry analysis of rhGH administration altered energy metabolism and steroidogenesis. Talanta 2024; 266:125069. [PMID: 37574608 DOI: 10.1016/j.talanta.2023.125069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/05/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
Growth hormone, as a proteohormone, is primarily known of its dramatic effect on longitudinal growth. Recombinant DNA technology has provided a safe, abundant and comparatively cheap supply of human GH for growth hormone-deficient individuals. However, many healthy subjects, especially athletics, administrate GH for enhanced athletic performance or strength. A better and more comprehensive understanding of rhGH effect in healthy individuals is urgent and essential. In this study, we recruited 14 healthy young male and injected rhGH once. Untargeted LC-MS metabolomics profiling of serum and urine was performed before and after the rhGH injection. The GH-induced dysregulation of energy related pathways, such as amino acid metabolism, nucleotide metabolism, glycolysis and TCA cycle, was revealed. Moreover, individuals supplemented with micro-doses of rhGH exhibited significantly changed urinary steroidal profiles, suggesting a role of rhGH in both energy metabolism and steroidogenesis. We expect that our results will be helpful to provide new evidence on the effects of rhGH injection and provide potential biomarkers for rhGH administration.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Chemistry, and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
| | - Wenqian Zhuang
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, 200000, China
| | - Yang Wang
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, 200000, China
| | - Xin Xu
- Research Institute for Doping Control, Shanghai University of Sport, Shanghai, 200000, China.
| | - Liang Qiao
- Department of Chemistry, and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China.
| |
Collapse
|
3
|
Pierucci F, Chirco A, Meacci E. Irisin Is Target of Sphingosine-1-Phosphate/Sphingosine-1-Phosphate Receptor-Mediated Signaling in Skeletal Muscle Cells. Int J Mol Sci 2023; 24:10548. [PMID: 37445724 DOI: 10.3390/ijms241310548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Irisin is a hormone-like myokine produced in abundance by skeletal muscle (SkM) in response to exercise. This myokine, identical in humans and mice, is involved in many signaling pathways related to metabolic processes. Despite much evidence on the regulators of irisin and the relevance of sphingolipids for SkM cell biology, the contribution of these latter bioactive lipids to the modulation of the myokine in SkM is missing. In particular, we have examined the potential involvement in irisin formation/release of sphingosine-1-phosphate (S1P), an interesting bioactive molecule able to act as an intracellular lipid mediator as well as a ligand of specific G-protein-coupled receptors (S1PR). We demonstrate the existence of distinct intracellular pools of S1P able to affect the expression of the irisin precursor FNDC. In addition, we establish the crucial role of the S1P/S1PR axis in irisin formation/release as well as the autocrine/paracrine effects of irisin on myoblast proliferation and myogenic differentiation. Altogether, these findings provide the first evidence for a functional crosstalk between the S1P/S1PR axis and irisin signaling, which may open new windows for potential therapeutic treatment of SkM dysfunctions.
Collapse
Affiliation(s)
- Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Firenze, Italy
| | - Antony Chirco
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Firenze, Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Firenze, Italy
| |
Collapse
|
4
|
Meacci E, Pierucci F, Garcia-Gil M. Skeletal Muscle and COVID-19: The Potential Involvement of Bioactive Sphingolipids. Biomedicines 2022; 10:biomedicines10051068. [PMID: 35625805 PMCID: PMC9138286 DOI: 10.3390/biomedicines10051068] [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: 04/04/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 virus infection is the cause of the coronavirus disease 2019 (COVID-19), which is still spreading over the world. The manifestation of this disease can range from mild to severe and can be limited in time (weeks) or persist for months in about 30–50% of patients. COVID-19 is considered a multiple organ dysfunction syndrome and the musculoskeletal system manifestations are beginning to be considered of absolute importance in both COVID-19 patients and in patients recovering from the SARS-CoV-2 infection. Musculoskeletal manifestations of COVID-19 and other coronavirus infections include loss of muscle mass, muscle weakness, fatigue or myalgia, and muscle injury. The molecular mechanisms by which SARS-CoV-2 can cause damage to skeletal muscle (SkM) cells are not yet well understood. Sphingolipids (SLs) represent an important class of eukaryotic lipids with structural functions as well as bioactive molecules able to modulate crucial processes, including inflammation and viral infection. In the last two decades, several reports have highlighted the role of SLs in modulating SkM cell differentiation, regeneration, aging, response to insulin, and contraction. This review summarizes the consequences of SARS-CoV-2 infection on SkM and the potential involvement of SLs in the tissue responses to virus infection. In particular, we highlight the role of sphingosine 1-phosphate signaling in order to aid the prediction of novel targets for preventing and/or treating acute and long-term musculoskeletal manifestations of virus infection in COVID-19.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Unit of Biochemical Sciences and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50121 Florence, Italy;
- Interuniversity Institute of Myology, University of Florence, 50121 Florence, Italy
- Correspondence: ; Tel.: +39-055-2751231
| | - Federica Pierucci
- Unit of Biochemical Sciences and Molecular Biology, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale GB Morgagni 50, 50121 Florence, Italy;
| | - Mercedes Garcia-Gil
- Unit of Physiology, Department of Biology, University of Pisa, Via S. Zeno 31, 56127 Pisa, Italy;
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| |
Collapse
|
5
|
Bernacchioni C, Squecco R, Gamberi T, Ghini V, Schumacher F, Mannelli M, Garella R, Idrizaj E, Cencetti F, Puliti E, Bruni P, Turano P, Fiaschi T, Donati C. S1P Signalling Axis Is Necessary for Adiponectin-Directed Regulation of Electrophysiological Properties and Oxidative Metabolism in C2C12 Myotubes. Cells 2022; 11:713. [PMID: 35203362 PMCID: PMC8869893 DOI: 10.3390/cells11040713] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Adiponectin (Adn), released by adipocytes and other cell types such as skeletal muscle, has insulin-sensitizing and anti-inflammatory properties. Sphingosine 1-phosphate (S1P) is reported to act as effector of diverse biological actions of Adn in different tissues. S1P is a bioactive sphingolipid synthesized by the phosphorylation of sphingosine catalyzed by sphingosine kinase (SK) 1 and 2. Consolidated findings support the key role of S1P in the biology of skeletal muscle. METHODS AND RESULTS Here we provide experimental evidence that S1P signalling is modulated by globular Adn treatment being able to increase the phosphorylation of SK1/2 as well as the mRNA expression levels of S1P4 in C2C12 myotubes. These findings were confirmed by LC-MS/MS that showed an increase of S1P levels after Adn treatment. Notably, the involvement of S1P axis in Adn action was highlighted since, when SK1 and 2 were inhibited by PF543 and ABC294640 inhibitors, respectively, not only the electrophysiological changes but also the increase of oxygen consumption and of aminoacid levels induced by the hormone, were significantly inhibited. CONCLUSION Altogether, these findings show that S1P biosynthesis is necessary for the electrophysiological properties and oxidative metabolism of Adn in skeletal muscle cells.
Collapse
Affiliation(s)
- Caterina Bernacchioni
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Roberta Squecco
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Veronica Ghini
- Magnetic Resonance Center (CERM), University of Florence, 50019 Florence, Italy; (V.G.); (P.T.)
| | - Fabian Schumacher
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, 14195 Berlin, Germany;
| | - Michele Mannelli
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Rachele Garella
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Eglantina Idrizaj
- Section of Physiological Sciences, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (R.S.); (R.G.); (E.I.)
| | - Francesca Cencetti
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Elisa Puliti
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Paola Bruni
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Paola Turano
- Magnetic Resonance Center (CERM), University of Florence, 50019 Florence, Italy; (V.G.); (P.T.)
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| | - Chiara Donati
- Department of Experimental and Clinical Biomedical Sciences “M. Serio”, University of Florence, 50134 Florence, Italy; (C.B.); (T.G.); (M.M.); (F.C.); (E.P.); (P.B.); (T.F.)
| |
Collapse
|
6
|
Cell instructive Liquid Crystalline Networks for myotube formation. iScience 2021; 24:103077. [PMID: 34568797 PMCID: PMC8449234 DOI: 10.1016/j.isci.2021.103077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/16/2021] [Accepted: 08/29/2021] [Indexed: 02/04/2023] Open
Abstract
Development of biological tissues in vitro is not a trivial task and requires the correct maturation of the selected cell line. To this aim, many attempts were done mainly by mimicking the biological environment using micro/nanopatterned or stimulated scaffolds. However, the obtainment of functional tissues in vitro is still far from being achieved. In contrast with the standard methods, we here present an easy approach for the maturation of myotubes toward the reproduction of muscular tissue. By using liquid crystalline networks with different stiffness and molecular alignment, we demonstrate how the material itself can give favorable interactions with myoblasts helping a correct differentiation. Electrophysiological studies demonstrate that myotubes obtained on these polymers have more adult-like morphology and better functional features with respect to those cultured on standard supports. The study opens to a platform for the differentiation of other cell lines in a simple and scalable way.
Collapse
|
7
|
Pierucci F, Frati A, Battistini C, Penna F, Costelli P, Meacci E. Control of Skeletal Muscle Atrophy Associated to Cancer or Corticosteroids by Ceramide Kinase. Cancers (Basel) 2021; 13:3285. [PMID: 34209043 PMCID: PMC8269416 DOI: 10.3390/cancers13133285] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 01/19/2023] Open
Abstract
Apart from cytokines and chemokines, sphingolipid mediators, particularly sphingosine-1-phosphate (S1P) and ceramide 1-phosphate (C1P), contribute to cancer and inflammation. Cancer, as well as other inflammatory conditions, are associated with skeletal muscle (SkM) atrophy, which is characterized by the unbalance between protein synthesis and degradation. Although the signaling pathways involved in SkM mass wasting are multiple, the regulatory role of simple sphingolipids is limited. Here, we report the impairment of ceramide kinase (CerK), the enzyme responsible for the phosphorylation of ceramide to C1P, associated with the accomplishment of atrophic phenotype in various experimental models of SkM atrophy: in vivo animal model bearing the C26 adenocarcinoma or Lewis lung carcinoma tumors, in human and murine SkM cells treated with the conditioned medium obtained from cancer cells or with the glucocorticoid dexamethasone. Notably, we demonstrate in all the three experimental approaches a drastic decrease of CerK expression. Gene silencing of CerK promotes the up-regulation of atrogin-1/MAFbx expression, which was also observed after cell treatment with C8-ceramide, a biologically active ceramide analogue. Conversely, C1P treatment significantly reduced the corticosteroid's effects. Altogether, these findings provide evidence that CerK, acting as a molecular modulator, may be a new possible target for SkM mass regulation associated with cancer or corticosteroids.
Collapse
Affiliation(s)
- Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| | - Chiara Battistini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, 10125 Torino, Italy; (F.P.); (P.C.)
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, 10125 Torino, Italy; (F.P.); (P.C.)
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”—Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy; (F.P.); (A.F.); (C.B.)
| |
Collapse
|
8
|
Rosa I, Marini M, Manetti M. Telocytes: An Emerging Component of Stem Cell Niche Microenvironment. J Histochem Cytochem 2021; 69:795-818. [PMID: 34165348 DOI: 10.1369/00221554211025489] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Telocytes (TCs) are newly identified interstitial cells characterized by thin and long cytoplasmic processes, called telopodes, which exhibit a distinctive moniliform shape and, often, a sinuous trajectory. Telopodes typically organize in intricate networks within the stromal space of most organs, where they communicate with neighboring cells by means of specialized cell-to-cell junctions or shedding extracellular vesicles. Hence, TCs are generally regarded as supporting cells that help in the maintenance of local tissue homeostasis, with an ever-increasing number of studies trying to explore their functions both in physiological and pathological conditions. Notably, TCs appear to be part of stem cell (SC) niches in different organs, including the intestine, skeletal muscle, heart, lung, and skin. Indeed, growing evidence points toward a possible implication of TCs in the regulation of the activity of tissue-resident SCs and in shaping the SC niche microenvironment, thus contributing to tissue renewal and repair. Here, we review how the introduction of TCs into the scientific literature has deepened our knowledge of the stromal architecture focusing on the intestine and skeletal muscle, two organs in which the recently unveiled unique relationship between TCs and SCs is currently in the spotlight as potential target for tissue regenerative purposes.
Collapse
Affiliation(s)
- Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mirca Marini
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
9
|
Bone Marrow-Mesenchymal Stromal Cell Secretome as Conditioned Medium Relieves Experimental Skeletal Muscle Damage Induced by Ex Vivo Eccentric Contraction. Int J Mol Sci 2021; 22:ijms22073645. [PMID: 33807453 PMCID: PMC8036477 DOI: 10.3390/ijms22073645] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Bone marrow-mesenchymal stem/stromal cells (MSCs) may offer promise for skeletal muscle repair/regeneration. Growing evidence suggests that the mechanisms underpinning the beneficial effects of such cells in muscle tissue reside in their ability to secrete bioactive molecules (secretome) with multiple actions. Hence, we examined the effects of MSC secretome as conditioned medium (MSC-CM) on ex vivo murine extensor digitorum longus muscle injured by forced eccentric contraction (EC). By combining morphological (light and confocal laser scanning microscopies) and electrophysiological analyses we demonstrated the capability of MSC-CM to attenuate EC-induced tissue structural damages and sarcolemnic functional properties’ modifications. MSC-CM was effective in protecting myofibers from apoptosis, as suggested by a reduced expression of pro-apoptotic markers, cytochrome c and activated caspase-3, along with an increase in the expression of pro-survival AKT factor. Notably, MSC-CM also reduced the EC-induced tissue redistribution and extension of telocytes/CD34+ stromal cells, distinctive cells proposed to play a “nursing” role for the muscle resident myogenic satellite cells (SCs), regarded as the main players of regeneration. Moreover, it affected SC functionality likely contributing to replenishment of the SC reservoir. This study provides the necessary groundwork for further investigation of the effects of MSC secretome in the setting of skeletal muscle injury and regenerative medicine.
Collapse
|
10
|
Hodun K, Chabowski A, Baranowski M. Sphingosine-1-phosphate in acute exercise and training. Scand J Med Sci Sports 2020; 31:945-955. [PMID: 33345415 DOI: 10.1111/sms.13907] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/27/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid found in all eukaryotic cells. Although it may function as an intracellular second messenger, most of its effects are induced extracellularly via activation of a family of five specific membrane receptors. Sphingosine-1-phosphate is enriched in plasma, where it is transported by high-density lipoprotein and albumin, as well as in erythrocytes and platelets which store and release large amounts of this sphingolipid. Sphingosine-1-phosphate regulates a host of cellular processes such as growth, proliferation, differentiation, migration, and apoptosis suppression. It was also shown to play an important role in skeletal muscle physiology and pathophysiology. In recent years, S1P metabolism in both muscle and blood was found to be modulated by exercise. In this review, we summarize the current knowledge on the effect of acute exercise and training on S1P metabolism, highlighting the role of this sphingolipid in skeletal muscle adaptation to physical effort.
Collapse
Affiliation(s)
- Katarzyna Hodun
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| | - Marcin Baranowski
- Department of Physiology, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
11
|
Squecco R, Chellini F, Idrizaj E, Tani A, Garella R, Pancani S, Pavan P, Bambi F, Zecchi-Orlandini S, Sassoli C. Platelet-Rich Plasma Modulates Gap Junction Functionality and Connexin 43 and 26 Expression During TGF-β1-Induced Fibroblast to Myofibroblast Transition: Clues for Counteracting Fibrosis. Cells 2020; 9:cells9051199. [PMID: 32408529 PMCID: PMC7290305 DOI: 10.3390/cells9051199] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/28/2020] [Accepted: 05/08/2020] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle repair/regeneration may benefit by Platelet-Rich Plasma (PRP) treatment owing to PRP pro-myogenic and anti-fibrotic effects. However, PRP anti-fibrotic action remains controversial. Here, we extended our previous researches on the inhibitory effects of PRP on in vitro transforming growth factor (TGF)-β1-induced differentiation of fibroblasts into myofibroblasts, the effector cells of fibrosis, focusing on gap junction (GJ) intercellular communication. The myofibroblastic phenotype was evaluated by cell shape analysis, confocal fluorescence microscopy and Western blotting analyses of α-smooth muscle actin and type-1 collagen expression, and electrophysiological recordings of resting membrane potential, resistance, and capacitance. PRP negatively regulated myofibroblast differentiation by modifying all the assessed parameters. Notably, myofibroblast pairs showed an increase of voltage-dependent GJ functionality paralleled by connexin (Cx) 43 expression increase. TGF-β1-treated cells, when exposed to a GJ blocker, or silenced for Cx43 expression, failed to differentiate towards myofibroblasts. Although a minority, myofibroblast pairs also showed not-voltage-dependent GJ currents and coherently Cx26 expression. PRP abolished the TGF-β1-induced voltage-dependent GJ current appearance while preventing Cx43 increase and promoting Cx26 expression. This study adds insights into molecular and functional mechanisms regulating fibroblast-myofibroblast transition and supports the anti-fibrotic potential of PRP, demonstrating the ability of this product to hamper myofibroblast generation targeting GJs.
Collapse
Affiliation(s)
- Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (R.S.); (E.I.); (R.G.)
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (R.S.); (E.I.); (R.G.)
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Rachele Garella
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, 50134 Florence, Italy; (R.S.); (E.I.); (R.G.)
| | - Sofia Pancani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Paola Pavan
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children’s Hospital, 50134 Florence, Italy; (P.P.); (F.B.)
| | - Franco Bambi
- Transfusion Medicine and Cell Therapy Unit, "A. Meyer" University Children’s Hospital, 50134 Florence, Italy; (P.P.); (F.B.)
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy; (F.C.); (A.T.); (S.P.); (S.Z.-O.)
- Correspondence: ; Tel.: +39-0552-7580-63
| |
Collapse
|
12
|
Tan-Chen S, Guitton J, Bourron O, Le Stunff H, Hajduch E. Sphingolipid Metabolism and Signaling in Skeletal Muscle: From Physiology to Physiopathology. Front Endocrinol (Lausanne) 2020; 11:491. [PMID: 32849282 PMCID: PMC7426366 DOI: 10.3389/fendo.2020.00491] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Sphingolipids represent one of the major classes of eukaryotic lipids. They play an essential structural role, especially in cell membranes where they also possess signaling properties and are capable of modulating multiple cell functions, such as apoptosis, cell proliferation, differentiation, and inflammation. Many sphingolipid derivatives, such as ceramide, sphingosine-1-phosphate, and ganglioside, have been shown to play many crucial roles in muscle under physiological and pathological conditions. This review will summarize our knowledge of sphingolipids and their effects on muscle fate, highlighting the role of this class of lipids in modulating muscle cell differentiation, regeneration, aging, response to insulin, and contraction. We show that modulating sphingolipid metabolism may be a novel and interesting way for preventing and/or treating several muscle-related diseases.
Collapse
Affiliation(s)
- Sophie Tan-Chen
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Jeanne Guitton
- Université Saclay, CNRS UMR 9197, Institut des Neurosciences Paris-Saclay, Orsay, France
| | - Olivier Bourron
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- Assistance Publique-Hôpitaux de Paris, Département de Diabétologie et Maladies Métaboliques, Hôpital Pitié-Salpêtrière, Paris, France
| | - Hervé Le Stunff
- Université Saclay, CNRS UMR 9197, Institut des Neurosciences Paris-Saclay, Orsay, France
| | - Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
- Institut Hospitalo-Universitaire ICAN, Paris, France
- *Correspondence: Eric Hajduch
| |
Collapse
|
13
|
S1P/S1P Receptor Signaling in Neuromuscolar Disorders. Int J Mol Sci 2019; 20:ijms20246364. [PMID: 31861214 PMCID: PMC6941007 DOI: 10.3390/ijms20246364] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/06/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022] Open
Abstract
The bioactive sphingolipid metabolite, sphingosine 1-phosphate (S1P), and the signaling pathways triggered by its binding to specific G protein-coupled receptors play a critical regulatory role in many pathophysiological processes, including skeletal muscle and nervous system degeneration. The signaling transduced by S1P binding appears to be much more complex than previously thought, with important implications for clinical applications and for personalized medicine. In particular, the understanding of S1P/S1P receptor signaling functions in specific compartmentalized locations of the cell is worthy of being better investigated, because in various circumstances it might be crucial for the development or/and the progression of neuromuscular diseases, such as Charcot-Marie-Tooth disease, myasthenia gravis, and Duchenne muscular dystrophy.
Collapse
|
14
|
Sphingosine 1-Phosphate (S1P)/ S1P Receptor Signaling and Mechanotransduction: Implications for Intrinsic Tissue Repair/Regeneration. Int J Mol Sci 2019; 20:ijms20225545. [PMID: 31703256 PMCID: PMC6888058 DOI: 10.3390/ijms20225545] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/16/2022] Open
Abstract
Tissue damage, irrespective from the underlying etiology, destroys tissue structure and, eventually, function. In attempt to achieve a morpho-functional recover of the damaged tissue, reparative/regenerative processes start in those tissues endowed with regenerative potential, mainly mediated by activated resident stem cells. These cells reside in a specialized niche that includes different components, cells and surrounding extracellular matrix (ECM), which, reciprocally interacting with stem cells, direct their cell behavior. Evidence suggests that ECM stiffness represents an instructive signal for the activation of stem cells sensing it by various mechanosensors, able to transduce mechanical cues into gene/protein expression responses. The actin cytoskeleton network dynamic acts as key mechanotransducer of ECM signal. The identification of signaling pathways influencing stem cell mechanobiology may offer therapeutic perspectives in the regenerative medicine field. Sphingosine 1-phosphate (S1P)/S1P receptor (S1PR) signaling, acting as modulator of ECM, ECM-cytoskeleton linking proteins and cytoskeleton dynamics appears a promising candidate. This review focuses on the current knowledge on the contribution of S1P/S1PR signaling in the control of mechanotransduction in stem/progenitor cells. The potential contribution of S1P/S1PR signaling in the mechanobiology of skeletal muscle stem cells will be argued based on the intriguing findings on S1P/S1PR action in this mechanically dynamic tissue.
Collapse
|
15
|
Manetti M, Tani A, Rosa I, Chellini F, Squecco R, Idrizaj E, Zecchi-Orlandini S, Ibba-Manneschi L, Sassoli C. Morphological evidence for telocytes as stromal cells supporting satellite cell activation in eccentric contraction-induced skeletal muscle injury. Sci Rep 2019; 9:14515. [PMID: 31601891 PMCID: PMC6787026 DOI: 10.1038/s41598-019-51078-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022] Open
Abstract
Although telocytes (TCs) have been proposed to play a “nursing” role in resident satellite cell (SC)-mediated skeletal muscle regeneration, currently there is no evidence of TC-SC morpho-functional interaction following tissue injury. Hence, we explored the presence of TCs and their relationship with SCs in an ex vivo model of eccentric contraction (EC)-induced muscle damage. EC-injured muscles showed structural/ultrastructural alterations and changes in electrophysiological sarcolemnic properties. TCs were identified in control and EC-injured muscles by either confocal immunofluorescence (i.e. CD34+CD31− TCs) or transmission electron microscopy (TEM). In EC-injured muscles, an extended interstitial network of CD34+ TCs/telopodes was detected around activated SCs displaying Pax7+ and MyoD+ nuclei. TEM revealed that TCs invaded the SC niche passing with their telopodes through a fragmented basal lamina and contacting the underlying activated SCs. TC-SC interaction after injury was confirmed in vitro by culturing single endomysial sheath-covered myofibers and sprouting TCs and SCs. EC-damaged muscle-derived TCs showed increased expression of the recognized pro-myogenic vascular endothelial growth factor-A, and SCs from the same samples exhibited increased MyoD expression and greater tendency to fuse into myotubes. Here, we provide the essential groundwork for further investigation of TC-SC interactions in the setting of skeletal muscle injury and regenerative medicine.
Collapse
Affiliation(s)
- Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy.
| | - Alessia Tani
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Roberta Squecco
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - Eglantina Idrizaj
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, University of Florence, Florence, Italy
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Lidia Ibba-Manneschi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Chiara Sassoli
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy.
| |
Collapse
|
16
|
Bittel DC, Jaiswal JK. Contribution of Extracellular Vesicles in Rebuilding Injured Muscles. Front Physiol 2019; 10:828. [PMID: 31379590 PMCID: PMC6658195 DOI: 10.3389/fphys.2019.00828] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/13/2019] [Indexed: 12/22/2022] Open
Abstract
Skeletal myofibers are injured due to mechanical stresses experienced during physical activity, or due to myofiber fragility caused by genetic diseases. The injured myofiber needs to be repaired or regenerated to restore the loss in muscle tissue function. Myofiber repair and regeneration requires coordinated action of various intercellular signaling factors-including proteins, inflammatory cytokines, miRNAs, and membrane lipids. It is increasingly being recognized release and transmission of these signaling factors involves extracellular vesicle (EV) released by myofibers and other cells in the injured muscle. Intercellular signaling by these EVs alters the phenotype of their target cells either by directly delivering the functional proteins and lipids or by modifying longer-term gene expression. These changes in the target cells activate downstream pathways involved in tissue homeostasis and repair. The EVs are heterogeneous with regards to their size, composition, cargo, location, as well as time-course of genesis and release. These differences impact on the subsequent repair and regeneration of injured skeletal muscles. This review focuses on how intracellular vesicle production, cargo packaging, and secretion by injured muscle, modulates specific reparative, and regenerative processes. Insights into the formation of these vesicles and their signaling properties offer new understandings of the orchestrated response necessary for optimal muscle repair and regeneration.
Collapse
Affiliation(s)
- Daniel C Bittel
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC, United States
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, Washington, DC, United States.,Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, United States
| |
Collapse
|
17
|
Editors T. Program and Book of the 15th Interuniversity Institute of Myology Meeting - Assisi (Italy), 2018. Eur J Transl Myol 2018; 28:7927. [PMID: 30662694 PMCID: PMC6317144 DOI: 10.4081/ejtm.2018.7927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 11/24/2022] Open
Abstract
Not available.
Collapse
Affiliation(s)
- The Editors
- Interuniversity Institute of Myology (IIM), Rome, Italy
| |
Collapse
|
18
|
Pierucci F, Frati A, Battistini C, Matteini F, Iachini MC, Vestri A, Penna F, Costelli P, Meacci E. Involvement of released sphingosine 1-phosphate/sphingosine 1-phosphate receptor axis in skeletal muscle atrophy. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3598-3614. [PMID: 30279138 DOI: 10.1016/j.bbadis.2018.08.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/27/2018] [Accepted: 08/30/2018] [Indexed: 12/18/2022]
Abstract
Skeletal muscle (SkM) atrophy is caused by several and heterogeneous conditions, such as cancer, neuromuscular disorders and aging. In most types of SkM atrophy overall rates of protein synthesis are suppressed, protein degradation is consistently elevated and atrogenes, such as the ubiquitin ligase Atrogin-1/MAFbx, are up-regulated. The molecular regulators of SkM waste are multiple and only in part known. Sphingolipids represent a class of bioactive molecules capable of modulating the destiny of many cell types, including SkM cells. In particular, we and others have shown that sphingosine 1phosphate (S1P), formed by sphingosine kinase (SphK), is able to act as trophic and morphogenic factor in myoblasts. Here, we report the first evidence that the atrophic phenotype observed in both muscle obtained from mice bearing the C26 adenocarcinoma and C2C12 myotubes treated with dexamethasone was characterized by reduced levels of active phospho-SphK1. The importance of SphK1 activity is also confirmed by the specific pharmacological inhibition of SphK1 able to increase Atrogin-1/MAFbx expression and reduce myotube size and myonuclei number. Furthermore, we found that SkM atrophy was accomplished by significant increase of S1P transporter Spns2 and in changes in the pattern of S1P receptor (S1PRs) subtype expression paralleled by increased Atrogin-1/MAFbx expression, suggesting a role for the released S1P and of specific S1PR-mediated signaling pathways in the control of the ubiquitin ligase. Altogether, these findings provide the first evidence that SphK1/released S1P/S1PR axis acts as a molecular regulator of SkM atrophy, thereby representing a new possible target for therapy in many patho-physiological conditions.
Collapse
Affiliation(s)
- Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Alessia Frati
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Chiara Battistini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Francesca Matteini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Maria Chiara Iachini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Ambra Vestri
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano (TO), Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Regione Gonzole 10, Orbassano (TO), Italy
| | - Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio" -Unit of Biochemical Sciences and Molecular Biology, University of Florence, Viale GB Morgagni, 50, Florence, Italy.
| |
Collapse
|
19
|
Rahar B, Chawla S, Pandey S, Bhatt AN, Saxena S. Sphingosine-1-phosphate pretreatment amends hypoxia-induced metabolic dysfunction and impairment of myogenic potential in differentiating C2C12 myoblasts by stimulating viability, calcium homeostasis and energy generation. J Physiol Sci 2018; 68:137-151. [PMID: 28070865 PMCID: PMC10717551 DOI: 10.1007/s12576-016-0518-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/26/2016] [Indexed: 12/19/2022]
Abstract
Sphingosine-1-phosphate (S1P) has a role in transpiration in patho-physiological signaling in skeletal muscles. The present study evaluated the pre-conditioning efficacy of S1P in facilitating differentiation of C2C12 myoblasts under a normoxic/hypoxic cell culture environment. Under normoxia, exogenous S1P significantly promoted C2C12 differentiation as evident from morphometric descriptors and differentiation markers of the mature myotubes, but it could facilitate only partial recovery from hypoxia-induced compromised differentiation. Pretreatment of S1P optimized the myokine secretion, intracellular calcium release and energy generation by boosting the aerobic/anaerobic metabolism and mitochondrial mass. In the hypoxia-exposed cells, there was derangement of the S1PR1-3 expression patterns, while the same could be largely restored with S1P pretreatment. This is being proposed as a plausible underlying mechanism for the observed pro-myogenic efficacy of exogenous S1P preconditioning. The present findings are an invaluable addition to the existing knowledge on the pro-myogenic potential of S1P and may prove beneficial in the field of hypoxia-related myo-pathologies.
Collapse
Affiliation(s)
- Babita Rahar
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Sonam Chawla
- Experimental Biology Division, Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Sanjay Pandey
- Division of Metabolic and Cell Signaling Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Anant Narayan Bhatt
- Division of Metabolic and Cell Signaling Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K. Mazumdar Road, Delhi, 110054, India
| | - Shweta Saxena
- Medicinal and Aromatic Plant Division, Defence Institute of High Altitude Research (DIHAR), Defence Research and Development Organization (DRDO), Ministry of Defence, Leh-Ladakh, 194101, Jammu and Kashmir, India.
| |
Collapse
|
20
|
Combined use of bone marrow-derived mesenchymal stromal cells (BM-MSCs) and platelet rich plasma (PRP) stimulates proliferation and differentiation of myoblasts in vitro: new therapeutic perspectives for skeletal muscle repair/regeneration. Cell Tissue Res 2018; 372:549-570. [PMID: 29404727 DOI: 10.1007/s00441-018-2792-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/06/2018] [Indexed: 10/18/2022]
Abstract
Satellite cell-mediated skeletal muscle repair/regeneration is compromised in cases of extended damage. Bone marrow mesenchymal stromal cells (BM-MSCs) hold promise for muscle healing but some criticisms hamper their clinical application, including the need to avoid animal serum contamination for expansion and the scarce survival after transplant. In this context, platelet-rich plasma (PRP) could offer advantages. Here, we compare the effects of PRP or standard culture media on C2C12 myoblast, satellite cell and BM-MSC viability, survival, proliferation and myogenic differentiation and evaluate PRP/BM-MSC combination effects in promoting myogenic differentiation. PRP induced an increase of mitochondrial activity and Ki67 expression comparable or even greater than that elicited by standard media and promoted AKT signaling activation in myoblasts and BM-MSCs and Notch-1 pathway activation in BM-MSCs. It stimulated MyoD, myogenin, α-sarcomeric actin and MMP-2 expression in myoblasts and satellite cell activation. Notably, PRP/BM-MSC combination was more effective than PRP alone. We found that BM-MSCs influenced myoblast responses through a paracrine activation of AKT signaling, contributing to shed light on BM-MSC action mechanisms. Our results suggest that PRP represents a good serum substitute for BM-MSC manipulation in vitro and could be beneficial towards transplanted cells in vivo. Moreover, it might influence muscle resident progenitors' fate, thus favoring the endogenous repair/regeneration mechanisms. Finally, within the limitations of an in vitro experimentation, this study provides an experimental background for considering the PRP/BM-MSC combination as a potential therapeutic tool for skeletal muscle damage, combining the beneficial effects of BM-MSCs and PRP on muscle tissue, while potentiating BM-MSC functionality.
Collapse
|
21
|
Sukocheva OA. Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming. Int J Mol Sci 2018; 19:ijms19020420. [PMID: 29385066 PMCID: PMC5855642 DOI: 10.3390/ijms19020420] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 02/05/2023] Open
Abstract
Sphingolipids, sphingolipid metabolizing enzymes, and their receptors network are being recognized as part of the signaling mechanisms, which govern breast cancer cell growth, migration, and survival during chemotherapy treatment. Approximately 70% of breast cancers are estrogen receptor (ER) positive and, thus, rely on estrogen signaling. Estrogen activates an intracellular network composed of many cytoplasmic and nuclear mediators. Some estrogen effects can be mediated by sphingolipids. Estrogen activates sphingosine kinase 1 (SphK1) and amplifies the intracellular concentration of sphingosine-1-phosphate (S1P) in breast cancer cells during stimulation of proliferation and survival. Specifically, Estrogen activates S1P receptors (S1PR) and induces growth factor receptor transactivation. SphK, S1P, and S1PR expression are causally associated with endocrine resistance and progression to advanced tumor stages in ER-positive breast cancers in vivo. Recently, the network of SphK/S1PR was shown to promote the development of ER-negative cancers and breast cancer stem cells, as well as stimulating angiogenesis. Novel findings confirm and broaden our knowledge about the cross-talk between sphingolipids and estrogen network in normal and malignant cells. Current S1PRs therapeutic inhibition was indicated as a promising chemotherapy approach in non-responsive and advanced malignancies. Considering that sphingolipid signaling has a prominent role in terminally differentiated cells, the impact should be considered when designing specific SphK/S1PR inhibitors. This study analyzes the dynamic of the transformation of sphingolipid axis during a transition from normal to pathological condition on the level of the whole organism. The sphingolipid-based mediation and facilitation of global effects of estrogen were critically accented as a bridging mechanism that should be explored in cancer prevention.
Collapse
Affiliation(s)
- Olga A Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| |
Collapse
|
22
|
Tsui JH, Janebodin K, Ieronimakis N, Yama DMP, Yang HS, Chavanachat R, Hays AL, Lee H, Reyes M, Kim DH. Harnessing Sphingosine-1-Phosphate Signaling and Nanotopographical Cues To Regulate Skeletal Muscle Maturation and Vascularization. ACS NANO 2017; 11:11954-11968. [PMID: 29156133 PMCID: PMC6133580 DOI: 10.1021/acsnano.7b00186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Despite possessing substantial regenerative capacity, skeletal muscle can suffer from loss of function due to catastrophic traumatic injury or degenerative disease. In such cases, engineered tissue grafts hold the potential to restore function and improve patient quality of life. Requirements for successful integration of engineered tissue grafts with the host musculature include cell alignment that mimics host tissue architecture and directional functionality, as well as vascularization to ensure tissue survival. Here, we have developed biomimetic nanopatterned poly(lactic-co-glycolic acid) substrates conjugated with sphingosine-1-phosphate (S1P), a potent angiogenic and myogenic factor, to enhance myoblast and endothelial maturation. Primary muscle cells cultured on these functionalized S1P nanopatterned substrates developed a highly aligned and elongated morphology and exhibited higher expression levels of myosin heavy chain, in addition to genes characteristic of mature skeletal muscle. We also found that S1P enhanced angiogenic potential in these cultures, as evidenced by elevated expression of endothelial-related genes. Computational analyses of live-cell videos showed a significantly improved functionality of tissues cultured on S1P-functionalized nanopatterns as indicated by greater myotube contraction displacements and velocities. In summary, our study demonstrates that biomimetic nanotopography and S1P can be combined to synergistically regulate the maturation and vascularization of engineered skeletal muscles.
Collapse
Affiliation(s)
- Jonathan H. Tsui
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Kajohnkiart Janebodin
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Anatomy, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Nicholas Ieronimakis
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, Washington, USA
| | - David M. P. Yama
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Hee Seok Yang
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, South Korea
| | | | - Aislinn L. Hays
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Haeshin Lee
- Department of Chemistry and the Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Morayma Reyes
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
23
|
Yano M, Minegishi Y, Sugita S, Ota N. Milk fat globule membrane supplementation with voluntary running exercise attenuates age-related motor dysfunction by suppressing neuromuscular junction abnormalities in mice. Exp Gerontol 2017; 97:29-37. [PMID: 28729214 DOI: 10.1016/j.exger.2017.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 07/05/2017] [Accepted: 07/17/2017] [Indexed: 11/28/2022]
Abstract
Age-related loss of skeletal muscle mass and function attenuates physical performance, and maintaining fine muscle innervation is known to play an important role in its prevention. We had previously shown that consumption of milk fat globule membrane (MFGM) with habitual exercise improves the muscle mass and motor function in humans and mice. Improvement of neuromuscular junction (NMJ) was suggested as one of the mechanisms underlying these effects. In this study, we evaluated the effect of MFGM intake combined with voluntary running (MFGM-VR) on morphological changes of NMJ and motor function in aging mice. Seven months following the intervention, the MFGM-VR group showed a significantly improved motor coordination in the rotarod test and muscle force in the grip strength test compared with the control group at 13 and 14months of age, respectively. In 14-month old control mice, the extensor digitorum longus muscle showed increased abnormal NMJs, such as fragmentation and denervation, compared with 6-month old young mice. However, such age-related deteriorations of NMJs were significantly suppressed in the MFGM-VR group. Increase in the expression of NMJ formation-related genes, such as agrin and LDL Receptor Related Protein 4 (LRP4), might contribute to this beneficial effect. Rotarod performance and grip strength showed significant negative correlation with the status of denervation and fragmentation of NMJs. These results suggest that MFGM intake with voluntary running exercise effectively suppresses age-related morphological deterioration of NMJ, thus contributing to improvement of motor function.
Collapse
Affiliation(s)
- Michiko Yano
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi 321-3497, Japan
| | - Yoshihiko Minegishi
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi 321-3497, Japan
| | - Satoshi Sugita
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi 321-3497, Japan
| | - Noriyasu Ota
- Biological Science Research, Kao Corporation, 2606 Akabane, Ichikai-machi, Haga-gun, Tochigi 321-3497, Japan.
| |
Collapse
|
24
|
Dietary supplementation with bovine-derived milk fat globule membrane lipids promotes neuromuscular development in growing rats. Nutr Metab (Lond) 2017; 14:9. [PMID: 28127382 PMCID: PMC5259894 DOI: 10.1186/s12986-017-0161-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 01/15/2017] [Indexed: 12/11/2022] Open
Abstract
Background The milk fat globule membrane (MFGM) is primarily composed of polar phospho- and sphingolipids, which have established biological effects on neuroplasticity. The present study aimed to investigate the effect of dietary MFGM supplementation on the neuromuscular system during post-natal development. Methods Growing rats received dietary supplementation with bovine-derived MFGM mixtures consisting of complex milk lipids (CML), beta serum concentrate (BSC) or a complex milk lipid concentrate (CMLc) (which lacks MFGM proteins) from post-natal day 10 to day 70. Results Supplementation with MFGM mixtures enriched in polar lipids (BSC and CMLc, but not CML) increased the plasma phosphatidylcholine (PC) concentration, with no effect on plasma phosphatidylinositol (PI), phosphatidylethanolamine (PE), phosphatidylserine (PS) or sphingomyelin (SM). In contrast, muscle PC was reduced in rats receiving supplementation with both BSC and CMLc, whereas muscle PI, PE, PS and SM remained unchanged. Rats receiving BSC and CMLc (but not CML) displayed a slow-to-fast muscle fibre type profile shift (MyHCI → MyHCIIa) that was associated with elevated expression of genes involved in myogenic differentiation (myogenic regulatory factors) and relatively fast fibre type specialisation (Myh2 and Nfatc4). Expression of neuromuscular development genes, including nerve cell markers, components of the synaptogenic agrin–LRP4 pathway and acetylcholine receptor subunits, was also increased in muscle of rats supplemented with BSC and CMLc (but not CML). Conclusions These findings demonstrate that dietary supplementation with bovine-derived MFGM mixtures enriched in polar lipids can promote neuromuscular development during post-natal growth in rats, leading to shifts in adult muscle phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s12986-017-0161-y) contains supplementary material, which is available to authorized users.
Collapse
|
25
|
Vitamin D 3 protects against Aβ peptide cytotoxicity in differentiated human neuroblastoma SH- SY5Y cells: A role for S1P1/p38MAPK/ATF4 axis. Neuropharmacology 2017; 116:328-342. [PMID: 28077289 DOI: 10.1016/j.neuropharm.2017.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 02/06/2023]
Abstract
Besides its classical function of bone metabolism regulation, 1alpha, 25-dihydroxyvitamin D3 (1,25(OH)2D3), acts on a variety of tissues including the nervous system, where the hormone plays an important role as neuroprotective, antiproliferating and differentiating agent. Sphingolipids are bioactive lipids that play critical and complex roles in regulating cell fate. In the present paper we have investigated whether sphingolipids are involved in the protective action of 1,25(OH)2D3. We have found that 1,25(OH)2D3 prevents amyloid-β peptide (Aβ(1-42)) cytotoxicity both in differentiated SH-SY5Y human neuroblastoma cells and in vivo. In differentiated SH-SY5Y cells, Aβ(1-42) strongly reduces the sphingosine-1-phosphate (S1P)/ceramide (Cer) ratio while 1,25(OH)2D3 partially reverts this effect. 1,25(OH)2D3 reverts also the Aβ(1-42)-induced reduction of sphingosine kinase activity. We have also studied the crosstalk between 1,25(OH)2D3 and S1P signaling pathways downstream to the activation of S1P receptor subtype S1P1. Notably, we found that 1,25(OH)2D3 prevents the reduction of S1P1 expression promoted by Aβ(1-42) and thereby it modulates the downstream signaling leading to ER stress damage (p38MAPK/ATF4). Similar effects were observed by using ZK191784. In addition, chronic treatment with 1,25(OH)2D3 protects from aggregated Aβ(1-42)-induced damage in the CA1 region of the rat hippocampus and promotes cell proliferation in the hippocampal dentate gyrus of adult mice. In conclusion, these results represent the first evidence of the role of 1,25(OH)2D3 and its structural analogue ZK191784 in counteracting the Aβ(1-42) peptide-induced toxicity through the modulation of S1P/S1P1/p38MAPK/ATF4 pathway in differentiated SH-SY5Y cells.
Collapse
|
26
|
Frati A, Ricci B, Pierucci F, Nistri S, Bani D, Meacci E. Role of sphingosine kinase/S1P axis in ECM remodeling of cardiac cells elicited by relaxin. Mol Endocrinol 2016; 29:53-67. [PMID: 25415609 DOI: 10.1210/me.2014-1201] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The initiation and progression of heart failure is linked to adverse cardiac remodeling of the extracellular matrix (ECM) during disease mainly through the deregulation of myocardial metalloproteinases (MMPs). Relaxin (RLX), a peptide hormone acting as a physiological cardiac effector, is a key regulator of ECM remodeling in reproductive and nonreproductive tissues. Studying primary cultures of mouse cardiac muscle cells and rat H9c2 cardiomyoblasts, we have obtained evidence for a new signaling pathway activated by RLX to induce ECM remodeling that involves the bioactive sphingolipids sphingosine-1-phosphate (S1P) and ceramide. In both cell populations, recombinant human RLX increased sphingosine kinase activity and S1P formation, whereas sphingomyelin and ceramide content were decreased in [(3)H]serine-labeled cells. According to the literature, RLX promoted MMP-2 and MMP-9 expression/release. Pharmacological inhibition of sphingolipid metabolism and silencing of sphingosine kinase 1, the enzyme responsible for S1P formation, were able to prevent MMP expression/release elicited by the hormone and induce the expression of tissue inhibitor of MMPs. In addition, we found that sphingolipid signaling is required for the regulation of connective tissue growth factor, a member of the CCN 1-3 family of genes that are involved in cell proliferation and differentiation. Finally, the induction of cardiomyoblast maturation induced by RLX was also found to be counteracted by inhibition of S1P formation. In conclusion, these findings provide a novel mechanism by which RLX acts on cardiac ECM remodeling and cardiac cell differentiation and offer interesting therapeutic options to prevent heart fibrosis and to favor myocardial regeneration.
Collapse
Affiliation(s)
- Alessia Frati
- Department of Biomedical, Experimental, and Clinical Sciences (A.F., B.R., F.P., E.M.), Research Unit of Biochemistry, and Department of Experimental and Clinical Medicine (S.N., D.B.), Research Unit of Histology and Embryology, University of Florence, 50134 Florence, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Frati A, Landi D, Marinelli C, Gianni G, Fontana L, Migliorini M, Pierucci F, Garcia-Gil M, Meacci E. Nutraceutical properties of chestnut flours: beneficial effects on skeletal muscle atrophy. Food Funct 2015; 5:2870-82. [PMID: 25183412 DOI: 10.1039/c4fo00353e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Plants contain a wide range of non-nutritive phytochemicals, many of which have protective or preventive properties for human diseases. The aim of the present work has been to investigate the nutraceutical properties of sweet chestnut flour extracts obtained from fruits collected from 7 geographic areas of Tuscany (Italy), and their ability in modulating skeletal muscle atrophy. We found that the cultivars from different geographic areas are characterized by the composition and quantity of various nutrients and specific bioactive components, such as tocopherols, polyphenols and sphingolipids. The nutraceutical properties of chestnut sweet flours have been evaluated in C2C12 myotubes induced to atrophy by serum deprivation or dexamethasone. We found that the pretreatment with both total extracts of tocopherols and sphingolipids is able to counterbalance cell atrophy, reducing the decrease in myotube size and myonuclei number, and attenuating protein degradation and the increase in expression of MAFbx/atrogin-1 (a muscle-specific atrophy marker). By contrast, polyphenol extracts were not able to prevent atrophy. Since we also found that γ-tocopherol is the major form of tocopherol in sweet flour and its content differs depending on the procedure of sweet flour preparation, the mechanisms by which γ-tocopherol as well as sphingolipids affect skeletal muscle cell atrophy have been also investigated. This is the first evidence that chestnut sweet flour is a natural source of specific bioactive components with a relevant role in the prevention of cell degeneration and maintenance of skeletal muscle mass, opening important implications in designing appropriate nutritional therapeutic approaches to skeletal muscle atrophy.
Collapse
Affiliation(s)
- Alessia Frati
- Dept. of Experimental Clinical and Biomedical Sciences, Unit of Biochemical Sciences, University of Florence, Viale GB Morgagni 50, 50134 Florence, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Giacomello E, Quarta M, Paolini C, Squecco R, Fusco P, Toniolo L, Blaauw B, Formoso L, Rossi D, Birkenmeier C, Peters LL, Francini F, Protasi F, Reggiani C, Sorrentino V. Deletion of small ankyrin 1 (sAnk1) isoforms results in structural and functional alterations in aging skeletal muscle fibers. Am J Physiol Cell Physiol 2014; 308:C123-38. [PMID: 25354526 DOI: 10.1152/ajpcell.00090.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Muscle-specific ankyrins 1 (sAnk1) are a group of small ankyrin 1 isoforms, of which sAnk1.5 is the most abundant. sAnk1 are localized in the sarcoplasmic reticulum (SR) membrane from where they interact with obscurin, a myofibrillar protein. This interaction appears to contribute to stabilize the SR close to the myofibrils. Here we report the structural and functional characterization of skeletal muscles from sAnk1 knockout mice (KO). Deletion of sAnk1 did not change the expression and localization of SR proteins in 4- to 6-mo-old sAnk1 KO mice. Structurally, the main modification observed in skeletal muscles of adult sAnk1 KO mice (4-6 mo of age) was the reduction of SR volume at the sarcomere A band level. With increasing age (at 12-15 mo of age) extensor digitorum longus (EDL) skeletal muscles of sAnk1 KO mice develop prematurely large tubular aggregates, whereas diaphragm undergoes significant structural damage. Parallel functional studies revealed specific changes in the contractile performance of muscles from sAnk1 KO mice and a reduced exercise tolerance in an endurance test on treadmill compared with control mice. Moreover, reduced Qγ charge and L-type Ca(2+) current, which are indexes of affected excitation-contraction coupling, were observed in diaphragm fibers from 12- to 15-mo-old mice, but not in other skeletal muscles from sAnk1 KO mice. Altogether, these findings show that the ablation of sAnk1, by altering the organization of the SR, renders skeletal muscles susceptible to undergo structural and functional alterations more evident with age, and point to an important contribution of sAnk1 to the maintenance of the longitudinal SR architecture.
Collapse
Affiliation(s)
- E Giacomello
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy; IIM-Interuniversity Institute of Myology
| | - M Quarta
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - C Paolini
- Ce.S.I., Center for Research on Ageing and Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio, Chieti, Italy; IIM-Interuniversity Institute of Myology
| | - R Squecco
- Department of Experimental and Clinical Medicine, University of Florence, Florence Italy; IIM-Interuniversity Institute of Myology
| | - P Fusco
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - L Toniolo
- Department of Biomedical Sciences, University of Padova, Padua, Italy; IIM-Interuniversity Institute of Myology
| | - B Blaauw
- Department of Biomedical Sciences, University of Padova, Padua, Italy; IIM-Interuniversity Institute of Myology; Venetian Institute of Molecular Medicine, Padua, Italy
| | - L Formoso
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - D Rossi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy; IIM-Interuniversity Institute of Myology
| | | | | | - F Francini
- Department of Experimental and Clinical Medicine, University of Florence, Florence Italy; IIM-Interuniversity Institute of Myology
| | - F Protasi
- Ce.S.I., Center for Research on Ageing and Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio, Chieti, Italy; IIM-Interuniversity Institute of Myology
| | - C Reggiani
- Department of Biomedical Sciences, University of Padova, Padua, Italy; IIM-Interuniversity Institute of Myology; CNR-Neuroscience Institute, Padua, Italy; and
| | - V Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy; IIM-Interuniversity Institute of Myology;
| |
Collapse
|
29
|
An endoplasmic reticulum stress-initiated sphingolipid metabolite, ceramide-1-phosphate, regulates epithelial innate immunity by stimulating β-defensin production. Mol Cell Biol 2014; 34:4368-78. [PMID: 25312644 DOI: 10.1128/mcb.00599-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Antimicrobial peptides (AMP) are ubiquitous innate immune elements in epithelial tissues. We recently discovered that a signaling lipid, the ceramide metabolite sphingosine-1-phosphate (S1P), regulates production of a major AMP, cathelicidin antimicrobial peptide (CAMP), in response to a subtoxic level of endoplasmic reticulum (ER) stress that can be induced by external perturbants in keratinocytes. We hypothesized that an ER stress-initiated signal could also regulate production of another major class of AMPs: i.e., the human beta-defensins 2 (hBD2) and 3 (hBD3). Keratinocytes stimulated with a pharmacological ER stressor, thapsigargin (Tg), increased hBD2/hBD3 as well as CAMP mRNA expression. While inhibition of sphingosine-1-phosphate production did not alter hBD expression following ER stress, blockade of ceramide-1-phosphate (C1P) suppressed Tg-induced hBD2/hBD3 but not CAMP expression. Exogenous C1P also increased hBD2/hBD3 production, indicating that C1P stimulates hBD expression. We showed further that C1P-induced hBD2/hBD3 expression is regulated by a novel pathway in which C1P stimulates downstream hBD via a cPLA2a→15d-PGJ2→PPARα/PPARβ/δ→Src kinase→STAT1/STAT3 transcriptional mechanism. Finally, conditioned medium from C1P-stimulated keratinocytes showed antimicrobial activity against Staphylococcus aureus. In summary, our present and recent studies discovered two new regulatory mechanisms of key epidermal AMP, hBD2/hBD3 and CAMP. The C1P and S1P pathways both signal to enhance innate immunity in response to ER stress.
Collapse
|
30
|
Sassoli C, Frati A, Tani A, Anderloni G, Pierucci F, Matteini F, Chellini F, Zecchi Orlandini S, Formigli L, Meacci E. Mesenchymal stromal cell secreted sphingosine 1-phosphate (S1P) exerts a stimulatory effect on skeletal myoblast proliferation. PLoS One 2014; 9:e108662. [PMID: 25264785 PMCID: PMC4181304 DOI: 10.1371/journal.pone.0108662] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/25/2014] [Indexed: 12/20/2022] Open
Abstract
Bone-marrow-derived mesenchymal stromal cells (MSCs) have the potential to significantly contribute to skeletal muscle healing through the secretion of paracrine factors that support proliferation and enhance participation of the endogenous muscle stem cells in the process of repair/regeneration. However, MSC-derived trophic molecules have been poorly characterized. The aim of this study was to investigate paracrine signaling effects of MSCs on skeletal myoblasts. It was found, using a biochemical and morphological approach that sphingosine 1-phosphate (S1P), a natural bioactive lipid exerting a broad range of muscle cell responses, is secreted by MSCs and represents an important factor by which these cells exert their stimulatory effects on C2C12 myoblast and satellite cell proliferation. Indeed, exposure to conditioned medium obtained from MSCs cultured in the presence of the selective sphingosine kinase inhibitor (iSK), blocked increased cell proliferation caused by the conditioned medium from untreated MSCs, and the addition of exogenous S1P in the conditioned medium from MSCs pre-treated with iSK further increased myoblast proliferation. Finally, we also demonstrated that the myoblast response to MSC-secreted vascular endothelial growth factor (VEGF) involves the release of S1P from C2C12 cells. Our data may have important implications in the optimization of cell-based strategies to promote skeletal muscle regeneration.
Collapse
Affiliation(s)
- Chiara Sassoli
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Alessia Frati
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Alessia Tani
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Giulia Anderloni
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Federica Pierucci
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Francesca Matteini
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
| | - Flaminia Chellini
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Sandra Zecchi Orlandini
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Lucia Formigli
- Department of Experimental and Clinical Medicine - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Elisabetta Meacci
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” - Unit of Biochemical Sciences and Molecular Biology, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
31
|
Abstract
As stem cells (SCs) in adult organs continue to be identified and characterized, it becomes clear that their survival, quiescence, and activation depend on specific signals in their microenvironment, or niche. Although adult SCs of diverse tissues differ by their developmental origin, cycling activity, and regenerative capacity, there appear to be conserved similarities regarding the cellular and molecular components of the SC niche. Interestingly, many organs house both slow-cycling and fast-cycling SC populations, which rely on the coexistence of quiescent and inductive niches for proper regulation. In this review we present a general definition of adult SC niches in the most studied mammalian systems. We further focus on dissecting their cellular organization and on highlighting recently identified key molecular regulators. Finally, we detail the potential involvement of the SC niche in tissue degeneration, with a particular emphasis on aging and cancer.
Collapse
Affiliation(s)
- Amélie Rezza
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Rachel Sennett
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Michael Rendl
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
32
|
Chandran S, Cairns MT, O'Brien M, Smith TJ. Transcriptomic effects of estradiol treatment on cultured human uterine smooth muscle cells. Mol Cell Endocrinol 2014; 393:16-23. [PMID: 24942541 DOI: 10.1016/j.mce.2014.05.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/07/2014] [Accepted: 05/22/2014] [Indexed: 11/20/2022]
Abstract
Contractility of the myometrial smooth muscle cells during the estrous cycle and pregnancy is modulated by estrogen but the temporal expression of estrogen (relative to progesterone) and the type of contraction involved are distinctly different in pregnancy and estrous. This in vitro cell culture study investigated the global gene expression profile of human uterine smooth muscle cells (hUtSMCs) following 17β-estradiol (E2) treatment. In response to E2 treatment 540 genes, many of which have not been previously described as estrogen responsive, were identified as significantly differentially expressed. These genes are involved in biological processes that include muscle contraction, cell migration and adhesion, apoptosis and phosphorylation. Evidence from this study suggests that 17β-estradiol may have effects that are contrary to an overall contraction phenotype. The hUtSMC in vitro culture system is a useful model to investigate steroid effects on smooth muscle cells and may provide additional clues as to how smooth muscle cells behave in vivo.
Collapse
Affiliation(s)
- Sreenath Chandran
- National Centre for Biomedical Engineering Science, NUI Galway, Galway, Ireland
| | - Michael T Cairns
- National Centre for Biomedical Engineering Science, NUI Galway, Galway, Ireland
| | - Margaret O'Brien
- National Centre for Biomedical Engineering Science, NUI Galway, Galway, Ireland
| | - Terry J Smith
- National Centre for Biomedical Engineering Science, NUI Galway, Galway, Ireland.
| |
Collapse
|
33
|
Sassoli C, Nosi D, Tani A, Chellini F, Mazzanti B, Quercioli F, Zecchi-Orlandini S, Formigli L. Defining the role of mesenchymal stromal cells on the regulation of matrix metalloproteinases in skeletal muscle cells. Exp Cell Res 2014; 323:297-313. [PMID: 24631289 DOI: 10.1016/j.yexcr.2014.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 03/01/2014] [Accepted: 03/03/2014] [Indexed: 12/11/2022]
Abstract
Recent studies indicate that mesenchymal stromal cell (MSC) transplantation improves healing of injured and diseased skeletal muscle, although the mechanisms of benefit are poorly understood. In the present study, we investigated whether MSCs and/or their trophic factors were able to regulate matrix metalloproteinase (MMP) expression and activity in different cells of the muscle tissue. MSCs in co-culture with C2C12 cells or their conditioned medium (MSC-CM) up-regulated MMP-2 and MMP-9 expression and function in the myoblastic cells; these effects were concomitant with the down-regulation of the tissue inhibitor of metalloproteinases (TIMP)-1 and -2 and with increased cell motility. In the single muscle fiber experiments, MSC-CM administration increased MMP-2/9 expression in Pax-7(+) satellite cells and stimulated their mobilization, differentiation and fusion. The anti-fibrotic properties of MSC-CM involved also the regulation of MMPs by skeletal fibroblasts and the inhibition of their differentiation into myofibroblasts. The treatment with SB-3CT, a potent MMP inhibitor, prevented in these cells, the decrease of α-smooth actin and type-I collagen expression induced by MSC-CM, suggesting that MSC-CM could attenuate the fibrogenic response through mechanisms mediated by MMPs. Our results indicate that growth factors and cytokines released by these cells may modulate the fibrotic response and improve the endogenous mechanisms of muscle repair/regeneration.
Collapse
Affiliation(s)
- Chiara Sassoli
- Dept. of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - Daniele Nosi
- Dept. of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - Alessia Tani
- Dept. of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - Flaminia Chellini
- Dept. of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - Benedetta Mazzanti
- Dept. of Experimental and Clinical Medicine-Section of Haematology, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - Franco Quercioli
- CNR-National Institute of Optics (INO), Largo Enrico Fermi 6, 50125 Arcetri-Florence, Italy
| | - Sandra Zecchi-Orlandini
- Dept. of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy
| | - Lucia Formigli
- Dept. of Experimental and Clinical Medicine-Section of Anatomy and Histology, University of Florence, Largo Brambilla, 3, 50134, Florence, Italy.
| |
Collapse
|
34
|
de la Garza-Rodea AS, Baldwin DM, Oskouian B, Place RF, Bandhuvula P, Kumar A, Saba JD. Sphingosine phosphate lyase regulates myogenic differentiation via S1P receptor-mediated effects on myogenic microRNA expression. FASEB J 2013; 28:506-19. [PMID: 24158395 DOI: 10.1096/fj.13-233155] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
S1P lyase (SPL) catalyzes the irreversible degradation of sphingosine-1-phosphate (S1P), a bioactive lipid whose signaling activities regulate muscle differentiation, homeostasis, and satellite cell (SC) activation. By regulating S1P levels, SPL also controls SC recruitment and muscle regeneration, representing a potential therapeutic target for muscular dystrophy. We found that SPL is induced during myoblast differentiation. To investigate SPL's role in myogenesis at the cellular level, we generated and characterized a murine myoblast SPL-knockdown (SPL-KD) cell line lacking SPL. SPL-KD cells accumulated intracellular and extracellular S1P and failed to form myotubes under conditions that normally stimulate myogenic differentiation. Under differentiation conditions, SPL-KD cells also demonstrated delayed induction of 3 myogenic microRNAs (miRNAs), miR-1, miR-206, and miR-486. SPL-KD cells successfully differentiated when treated with an S1P1 agonist, S1P2 antagonist, and combination treatments, which also increased myogenic miRNA levels. SPL-KD cells transfected with mimics for miR-1 or miR-206 also overcame the differentiation block. Thus, we show for the first time that the S1P/SPL/S1P-receptor axis regulates the expression of a number of miRNAs, thereby contributing to myogenic differentiation.
Collapse
Affiliation(s)
- Anabel S de la Garza-Rodea
- 1Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA 94609, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Fortier M, Figeac N, White RB, Knopp P, Zammit PS. Sphingosine-1-phosphate receptor 3 influences cell cycle progression in muscle satellite cells. Dev Biol 2013; 382:504-16. [PMID: 23911934 PMCID: PMC3898928 DOI: 10.1016/j.ydbio.2013.07.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 07/06/2013] [Accepted: 07/11/2013] [Indexed: 12/17/2022]
Abstract
Skeletal muscle retains a resident stem cell population called satellite cells, which are mitotically quiescent in mature muscle, but can be activated to produce myoblast progeny for muscle homeostasis, hypertrophy and repair. We have previously shown that satellite cell activation is partially controlled by the bioactive phospholipid, sphingosine-1-phosphate, and that S1P biosynthesis is required for muscle regeneration. Here we investigate the role of sphingosine-1-phosphate receptor 3 (S1PR3) in regulating murine satellite cell function. S1PR3 levels were high in quiescent myogenic cells before falling during entry into cell cycle. Retrovirally-mediated constitutive expression of S1PR3 led to suppressed cell cycle progression in satellite cells, but did not overtly affect the myogenic program. Conversely, satellite cells isolated from S1PR3-null mice exhibited enhanced proliferation ex-vivo. In vivo, acute cardiotoxin-induced muscle regeneration was enhanced in S1PR3-null mice, with bigger muscle fibres compared to control mice. Importantly, genetically deleting S1PR3 in the mdx mouse model of Duchenne muscular dystrophy produced a less severe muscle dystrophic phenotype, than when signalling though S1PR3 was operational. In conclusion, signalling though S1PR3 suppresses cell cycle progression to regulate function in muscle satellite cells. Expression of S1PR3 is associated with non-cycling myoblasts. Constitutive expression of S1PR3 leads to reduced cell proliferation. Satellite cells lacking S1PR3 have enhanced proliferation. Muscle regeneration is improved in the absence of S1PR3. The dystrophic phenotype in mdx mice is improved by the absence of S1PR3.
Collapse
Affiliation(s)
- Mathieu Fortier
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | | | | | | | | |
Collapse
|
36
|
Stiber JA, Tang Y, Li T, Rosenberg PB. Cytoskeletal regulation of TRPC channels in the cardiorenal system. Curr Hypertens Rep 2013; 14:492-7. [PMID: 23054893 DOI: 10.1007/s11906-012-0313-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Transient receptor potential canonical (TRPC) channels have been implicated in several aspects of cardiorenal physiology including regulation of blood pressure, vasoreactivity, vascular remodeling, and glomerular filtration. Gain and loss of function studies also support the role of TRPC channels in adverse remodeling associated with cardiac hypertrophy and heart failure. This review discusses TRP channels in the cardiovascular and glomerular filtration systems and their role in disease pathogenesis. We describe the regulation of gating of TRPC channels in the cardiorenal system as well as the influence on activation of these channels by the underlying cytoskeleton and scaffolding proteins. We then focus on the role of TRP channels in the pathogenesis of adverse cardiac remodeling and as potential therapeutic targets in the treatment of heart failure.
Collapse
Affiliation(s)
- Jonathan A Stiber
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
37
|
Abstract
Adult skeletal muscle in mammals is a stable tissue under normal circumstances but has remarkable ability to repair after injury. Skeletal muscle regeneration is a highly orchestrated process involving the activation of various cellular and molecular responses. As skeletal muscle stem cells, satellite cells play an indispensible role in this process. The self-renewing proliferation of satellite cells not only maintains the stem cell population but also provides numerous myogenic cells, which proliferate, differentiate, fuse, and lead to new myofiber formation and reconstitution of a functional contractile apparatus. The complex behavior of satellite cells during skeletal muscle regeneration is tightly regulated through the dynamic interplay between intrinsic factors within satellite cells and extrinsic factors constituting the muscle stem cell niche/microenvironment. For the last half century, the advance of molecular biology, cell biology, and genetics has greatly improved our understanding of skeletal muscle biology. Here, we review some recent advances, with focuses on functions of satellite cells and their niche during the process of skeletal muscle regeneration.
Collapse
Affiliation(s)
- Hang Yin
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
38
|
Donati C, Cencetti F, Bruni P. New insights into the role of sphingosine 1-phosphate and lysophosphatidic acid in the regulation of skeletal muscle cell biology. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:176-84. [DOI: 10.1016/j.bbalip.2012.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/29/2012] [Accepted: 06/30/2012] [Indexed: 12/25/2022]
|
39
|
Abstract
Studies performed in the last fifteen years have clearly established that the bioactive sphingolipid sphingosine 1-phosphate (S1P) affects various different biological properties of myogenic precursor cells as well as physiological features of adult skeletal muscle. Noticeably, in myogenic precursor cells multiple growth factors and cytokines cross-communicate with S1P axis and the engagement of distinct S1P receptor subtypes appears to be crucially implicated in transmitting specific biological effects. This paper summarizes current research findings and discloses the potential for new therapeutics designed to alter S1P signaling with the aim of improving skeletal muscle repair.
Collapse
Affiliation(s)
- Paola Bruni
- Dipartimento di Scienze Biochimiche, Università di Firenze, Firenze, Italy.
| | | |
Collapse
|
40
|
Trophic actions of bone marrow-derived mesenchymal stromal cells for muscle repair/regeneration. Cells 2012; 1:832-50. [PMID: 24710532 PMCID: PMC3901134 DOI: 10.3390/cells1040832] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 09/28/2012] [Accepted: 10/09/2012] [Indexed: 12/30/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (BM-MSCs) represent the leading candidate cell in tissue engineering and regenerative medicine. These cells can be easily isolated, expanded in vitro and are capable of providing significant functional benefits after implantation in the damaged muscle tissues. Despite their plasticity, the participation of BM-MSCs to new muscle fiber formation is controversial; in fact, emerging evidence indicates that their therapeutic effects occur without signs of long-term tissue engraftment and involve the paracrine secretion of cytokines and growth factors with multiple effects on the injured tissue, including modulation of inflammation and immune reaction, positive extracellular matrix (ECM) remodeling, angiogenesis and protection from apoptosis. Recently, a new role for BM-MSCs in the stimulation of muscle progenitor cells proliferation has been demonstrated, suggesting the potential ability of these cells to influence the fate of local stem cells and augment the endogenous mechanisms of repair/regeneration in the damaged tissues.
Collapse
|
41
|
Bini F, Frati A, Garcia-Gil M, Battistini C, Granado M, Martinesi M, Mainardi M, Vannini E, Luzzati F, Caleo M, Peretto P, Gomez-Muñoz A, Meacci E. New signalling pathway involved in the anti-proliferative action of vitamin D3 and its analogues in human neuroblastoma cells. A role for ceramide kinase. Neuropharmacology 2012; 63:524-37. [DOI: 10.1016/j.neuropharm.2012.04.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 04/06/2012] [Accepted: 04/21/2012] [Indexed: 01/12/2023]
|
42
|
Sassoli C, Pini A, Chellini F, Mazzanti B, Nistri S, Nosi D, Saccardi R, Quercioli F, Zecchi-Orlandini S, Formigli L. Bone marrow mesenchymal stromal cells stimulate skeletal myoblast proliferation through the paracrine release of VEGF. PLoS One 2012; 7:e37512. [PMID: 22815682 PMCID: PMC3398011 DOI: 10.1371/journal.pone.0037512] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 04/24/2012] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are the leading cell candidates in the field of regenerative medicine. These cells have also been successfully used to improve skeletal muscle repair/regeneration; however, the mechanisms responsible for their beneficial effects remain to be clarified. On this basis, in the present study, we evaluated in a co-culture system, the ability of bone-marrow MSCs to influence C2C12 myoblast behavior and analyzed the cross-talk between the two cell types at the cellular and molecular level. We found that myoblast proliferation was greatly enhanced in the co-culture as judged by time lapse videomicroscopy, cyclin A expression and EdU incorporation. Moreover, myoblasts immunomagnetically separated from MSCs after co-culture expressed higher mRNA and protein levels of Notch-1, a key determinant of myoblast activation and proliferation, as compared with the single culture. Notch-1 intracellular domain and nuclear localization of Hes-1, a Notch-1 target gene, were also increased in the co-culture. Interestingly, the myoblastic response was mainly dependent on the paracrine release of vascular endothelial growth factor (VEGF) by MSCs. Indeed, the addition of MSC-derived conditioned medium (CM) to C2C12 cells yielded similar results as those observed in the co-culture and increased the phosphorylation and expression levels of VEGFR. The treatment with the selective pharmacological VEGFR inhibitor, KRN633, resulted in a marked attenuation of the receptor activation and concomitantly inhibited the effects of MSC-CM on C2C12 cell growth and Notch-1 signaling. In conclusion, this study provides novel evidence for a role of MSCs in stimulating myoblast cell proliferation and suggests that the functional interaction between the two cell types may be exploited for the development of new and more efficient cell-based skeletal muscle repair strategies.
Collapse
Affiliation(s)
- Chiara Sassoli
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Alessandro Pini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Flaminia Chellini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Benedetta Mazzanti
- Department of Hematology, Cord Blood Bank, Careggi Hospital, University of Florence, Florence, Italy
| | - Silvia Nistri
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Riccardo Saccardi
- Department of Hematology, Cord Blood Bank, Careggi Hospital, University of Florence, Florence, Italy
| | - Franco Quercioli
- National Institute of Optics (INO), National Research Council (CNR), Sesto Fiorentino, Florence, Italy
| | - Sandra Zecchi-Orlandini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Lucia Formigli
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
43
|
Sphingosine-1-phosphate enhances satellite cell activation in dystrophic muscles through a S1PR2/STAT3 signaling pathway. PLoS One 2012; 7:e37218. [PMID: 22606352 PMCID: PMC3351440 DOI: 10.1371/journal.pone.0037218] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 04/15/2012] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) activates a widely expressed family of G protein-coupled receptors, serves as a muscle trophic factor and activates muscle stem cells called satellite cells (SCs) through unknown mechanisms. Here we show that muscle injury induces dynamic changes in S1P signaling and metabolism in vivo. These changes include early and profound induction of the gene encoding the S1P biosynthetic enzyme SphK1, followed by induction of the catabolic enzyme sphingosine phosphate lyase (SPL) 3 days later. These changes correlate with a transient increase in circulating S1P levels after muscle injury. We show a specific requirement for SphK1 to support efficient muscle regeneration and SC proliferation and differentiation. Mdx mice, which serve as a model for muscular dystrophy (MD), were found to be S1P-deficient and exhibited muscle SPL upregulation, suggesting that S1P catabolism is enhanced in dystrophic muscle. Pharmacological SPL inhibition increased muscle S1P levels, improved mdx muscle regeneration and enhanced SC proliferation via S1P receptor 2 (S1PR2)-dependent inhibition of Rac1, thereby activating Signal Transducer and Activator of Transcription 3 (STAT3), a central player in inflammatory signaling. STAT3 activation resulted in p21 and p27 downregulation in a S1PR2-dependent fashion in myoblasts. Our findings suggest that S1P promotes SC progression through the cell cycle by repression of cell cycle inhibitors via S1PR2/STAT3-dependent signaling and that SPL inhibition may provide a therapeutic strategy for MD.
Collapse
|
44
|
Hara M, Tabata K, Suzuki T, Do MKQ, Mizunoya W, Nakamura M, Nishimura S, Tabata S, Ikeuchi Y, Sunagawa K, Anderson JE, Allen RE, Tatsumi R. Calcium influx through a possible coupling of cation channels impacts skeletal muscle satellite cell activation in response to mechanical stretch. Am J Physiol Cell Physiol 2012; 302:C1741-50. [PMID: 22460715 DOI: 10.1152/ajpcell.00068.2012] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
When skeletal muscle is stretched or injured, satellite cells, resident myogenic stem cells positioned beneath the basal lamina of mature muscle fibers, are activated to enter the cell cycle. This signaling pathway is a cascade of events including calcium-calmodulin formation, nitric oxide (NO) radical production by NO synthase, matrix metalloproteinase activation, release of hepatocyte growth factor (HGF) from the extracellular matrix, and presentation of HGF to the receptor c-met, as demonstrated by assays of primary cultures and in vivo experiments. Here, we add evidence that two ion channels, the mechanosensitive cation channel (MS channel) and the long-lasting-type voltage-gated calcium-ion channel (L-VGC channel), mediate the influx of extracellular calcium ions in response to cyclic stretch in satellite cell cultures. When applied to 1-h stretch cultures with individual inhibitors for MS and L-VGC channels (GsMTx-4 and nifedipine, respectively) or with a less specific inhibitor (gadolinium chloride, Gd), satellite cell activation and upstream HGF release were abolished, as revealed by bromodeoxyuridine-incorporation assays and Western blotting of conditioned media, respectively. The inhibition was dose dependent with a maximum at 0.1 μM (GsMTx-4), 10 μM (nifedipine), or 100 μM (Gd) and canceled by addition of HGF to the culture media; a potent inhibitor for transient-type VGC channels (NNC55-0396, 100 μM) did not show any significant inhibitory effect. The stretch response was also abolished when calcium-chelator EGTA (1.8 mM) was added to the medium, indicating the significance of extracellular free calcium ions in our present activation model. Finally, cation/calcium channel dependencies were further documented by calcium-imaging analyses on stretched cells; results clearly demonstrated that calcium ion influx was abolished by GsMTx-4, nifedipine, and EGTA. Therefore, these results provide an additional insight that calcium ions may flow in through L-VGC channels by possible coupling with adjacent MS channel gating that promotes the local depolarization of cell membranes to initiate the satellite cell activation cascade.
Collapse
Affiliation(s)
- Minako Hara
- Department of Animal and Marine Bioresource Sciences, Graduate School of Agriculture, Kyushu University, Hakozaki, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid regulator of numerous important physiological and pathological processes in mammalian and nonmammalian cells. There are emerging evidence that many cell types can produce and release S1P; therefore, the quantification of its intracellular and extracellular content as well as the activity of sphingosine kinase (SphK), the enzyme responsible of S1P synthesis, is crucial to attribute to the SphK/S1P axis a functional significance in response to many different stimuli and in physiopathological conditions.This chapter describes experimental procedures to measure intracellular S1P formation in skeletal muscle cells and skeletal muscle fibers by using sphingolipid precursors. It also underlines the relevance of measuring S1P production in specific cellular compartments in order to attribute to S1P signaling a role in the biology of skeletal muscle cells.
Collapse
|
46
|
Calise S, Blescia S, Cencetti F, Bernacchioni C, Donati C, Bruni P. Sphingosine 1-phosphate stimulates proliferation and migration of satellite cells: role of S1P receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1823:439-50. [PMID: 22178384 DOI: 10.1016/j.bbamcr.2011.11.016] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 11/14/2011] [Accepted: 11/16/2011] [Indexed: 12/16/2022]
Abstract
Satellite cells are resident stem cells of skeletal muscle; they are normally quiescent but upon post-trauma activation start to proliferate and fuse with damaged fibers contributing to muscle regeneration. In this study the effect of the bioactive sphingolipid sphingosine 1-phosphate (S1P) on the proliferative and migratory response of murine satellite cells has been examined. S1P was found to stimulate labeled thymidine incorporation in a phosphatidylinositol 3-kinase-dependent manner. Moreover, by employing selective S1P receptor agonists and antagonists and silencing individual S1P receptors, the mitogenic action of S1P in satellite cells was shown to depend on S1P2 and S1P3. Notably, by using different experimental approaches S1P was found to positively influence satellite cell migration, necessary for their recruitment at the site of muscle damage. Interestingly, the specific silencing of individual S1P receptor subtypes demonstrated the pivotal role of S1P1 and S1P4 in mediating the S1P migratory effect. This latter result demonstrates for the first time that S1P4 receptor has a role in skeletal muscle cells, supporting the notion that this receptor subtype plays a biological action broader than that so far identified in lymphoid tissue. On the contrary, S1P2 was found to negatively regulate cell migration. Collectively, these results are in favour of an important function of S1P in satellite cell biology that could in principle be exploited as novel pharmacological target for improving skeletal muscle regeneration.
Collapse
Affiliation(s)
- Serena Calise
- Dipartimento di Scienze Biochimiche, Università di Firenze, 50134 Firenze, Italy
| | | | | | | | | | | |
Collapse
|