1
|
Guo L, Jin K, Sun Q, Zhang C, Chen X, Geng Z. Adiponectin regulates proliferation and differentiation of chicken skeletal muscle satellite cells via ERK1/2 and p38 signaling pathways. Poult Sci 2025; 104:104813. [PMID: 39823838 PMCID: PMC11786077 DOI: 10.1016/j.psj.2025.104813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025] Open
Abstract
Skeletal muscle satellite cells (SMSCs) are critical for postnatal skeletal muscle growth and regeneration. Adiponectin plays a pivotal role in regulating muscle glucose uptake and fatty acid metabolism. However, its function in the proliferation and differentiation of chicken SMSCs remains poorly understood. In this study, we investigated the effects of adiponectin on the proliferation and differentiation of in vitro cultured chicken SMSCs. Our results demonstrated that adiponectin promoted SMSCs proliferation while inhibiting myogenic differentiation and inducing adipogenic differentiation. RNA-seq analysis revealed enrichment of the MAPK signaling pathway, suggesting its potential involvement in the regulation of adiponectin on SMSCs activity. Western blot analysis revealed that adiponectin activated ERK1/2 phosphorylation and inhibited p38 phosphorylation during the process of the inhibition on myogenic differentiation in chicken SMSCs. Furthermore, suppression of ERK1/2 signaling with U0126 or activation of p38 signaling with SSK1 reversed the downregulated expression of myogenic differentiation marker MyHC, MyOD1, and MyOG induced by adiponectin. These findings validated that adiponectin impeded myogenic differentiation through activation of ERK1/2 and inhibition of p38 signaling pathways. Additionally, activation of p38 signaling pathway reduced the increased percentage of EdU-positive cells induced by adiponectin. Collectively, these findings demonstrated that adiponectin impedes myogenic differentiation of SMSCs through activating ERK1/2 and inhibiting p38 signaling pathways, while promoting proliferation by inhibiting p38 signaling pathway.
Collapse
Affiliation(s)
- Liping Guo
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, 130 Changjiang West Rd., Hefei 230036, PR China
| | - Kaiming Jin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Qi Sun
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Chenchao Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China
| | - Xiongyong Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, 130 Changjiang West Rd., Hefei 230036, PR China.
| | - Zhaoyu Geng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, PR China; Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, 130 Changjiang West Rd., Hefei 230036, PR China
| |
Collapse
|
2
|
Yoo E, Choi HJ, Han JY. Enhanced activation of signaling pathway by recombinant human adiponectin from genome-edited chickens. J Biotechnol 2024; 395:95-99. [PMID: 39341349 DOI: 10.1016/j.jbiotec.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/24/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
Adiponectin (ADPN) exerts various cellular and metabolic functions by activating signaling pathways, including extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) pathways, the protein kinase B (Akt) pathway, and the p38 mitogen-activated protein kinase (MAPK) pathway. However, generating functional recombinant human adiponectin (hADPN) in bacterial or mammalian cells is challenging. Although ADPN agonist peptides have been developed, problems like stability, solubility, and affinity for receptors remain. Recently, a genome-edited chicken bioreactor system was established, ensuring efficient ADPN production with optimal post-transcriptional modifications. We assessed the ability of egg white (EW)-derived hADPN, commercial hADPN, various ADPN agonist peptides, and globular ADPN on activation of the ERK1/2, Akt, and p38 MAPK pathways. EW-derived hADPN, abundant in hexamers and high molecular weight multimers, significantly phosphorylated ERK1/2 in serum-starved HEK293 cells after 15 min of treatment. Comparative analysis revealed that EW-derived hADPN and commercial hADPN induced greater phosphorylation of ERK1/2, Akt, and p38 MAPK than ADPN agonist peptides and globular ADPN, with EW-derived hADPN showing the highest activation. In summary, the finding that EW-derived hADPN strongly activates the ERK1/2, Akt, p38 MAPK signaling pathways highlights that an ADPN production system based on genome-edited chickens is an advantageous alternative to existing methods.
Collapse
Affiliation(s)
- Eunhui Yoo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hee Jung Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Yong Han
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
3
|
Massart IS, Kouakou A, Pelet N, Lause P, Schakman O, Loumaye A, Abou‐Samra M, Deldicque L, Bindels LB, Brichard SM, Thissen J. Administration of adiponectin receptor agonist AdipoRon relieves cancer cachexia by mitigating inflammation in tumour-bearing mice. J Cachexia Sarcopenia Muscle 2024; 15:919-933. [PMID: 38572511 PMCID: PMC11154773 DOI: 10.1002/jcsm.13454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 01/30/2024] [Accepted: 02/16/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Cancer cachexia is a life-threatening, inflammation-driven wasting syndrome that remains untreatable. Adiponectin, the most abundant adipokine, plays an important role in several metabolic processes as well as in inflammation modulation. Our aim was to test whether administration of AdipoRon (AR), a synthetic agonist of the adiponectin receptors, prevents the development of cancer cachexia and its related muscle atrophy. METHODS The effect of AR on cancer cachexia was investigated in two distinct murine models of colorectal cancer. First, 7-week-old CD2F1 male mice were subcutaneously injected with colon-26 carcinoma cells (C26) or vehicle (CT). Six days after injection, mice were treated for 5 days with AdipoRon (50 mg/kg/day; C26 + AR) or the corresponding vehicle (CT and C26). Additionally, a genetic model, the ApcMin/+ mouse, that develops spontaneously numerous intestinal polyps, was used. Eight-week-old male ApcMin/+ mice were treated with AdipoRon (50 mg/kg/day; Apc + AR) or the corresponding vehicle (Apc) over a period of 12 weeks, with C57BL/6J wild-type mice used as controls. In both models, several parameters were assessed in vivo: body weight, grip strength and serum parameters, as well as ex vivo: molecular changes in muscle, fat and liver. RESULTS The protective effect of AR on cachexia development was observed in both cachectic C26 and ApcMin/+ mice. In these mice, AR administration led to a significant alleviation of body weight loss and muscle wasting, together with rescued muscle strength (P < 0.05 for all). In both models, AR had a strong anti-inflammatory effect, reflected by lower systemic interleukin-6 levels (-55% vs. C26, P < 0.001 and -80% vs. Apc mice, P < 0.05), reduced muscular inflammation as indicated by lower levels of Socs3, phospho-STAT3 and Serpina3n, an acute phase reactant (P < 0.05 for all). In addition, AR blunted circulating levels of corticosterone (-46% vs. C26 mice, P < 0.001 and -60% vs. Apc mice, P < 0.05), the predominant murine glucocorticoid known to induce muscle atrophy. Accordingly, key glucocorticoid-responsive factors implicated in atrophy programmes were-or tended to be-significantly blunted in skeletal muscle by AR. Finally, AR protected against lipid metabolism alterations observed in ApcMin/+ mice, as it mitigated the increase in circulating triglyceride levels (-38%, P < 0.05) by attenuating hepatic triglyceride synthesis and fatty acid uptake by the liver. CONCLUSIONS Altogether, these results show that AdipoRon rescued the cachectic phenotype by alleviating body weight loss and muscle atrophy, along with restraining inflammation and hypercorticism in preclinical murine models. Therefore, AdipoRon could represent an innovative therapeutic strategy to counteract cancer cachexia.
Collapse
Affiliation(s)
- Isabelle S. Massart
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Axell‐Natalie Kouakou
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Nathan Pelet
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Pascale Lause
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Olivier Schakman
- Institute of NeuroscienceUniversité catholique de LouvainLouvain‐la‐NeuveBelgium
| | - Audrey Loumaye
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Michel Abou‐Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Louise Deldicque
- Institute of NeuroscienceUniversité catholique de LouvainLouvain‐la‐NeuveBelgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research InstituteUniversité catholique de LouvainBrusselsBelgium
| | - Sonia M. Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| | - Jean‐Paul Thissen
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical ResearchUniversité catholique de LouvainBrusselsBelgium
| |
Collapse
|
4
|
Neri S, Assirelli E, Manzetti M, Viroli G, Ialuna M, Traversari M, Ciaffi J, Ursini F, Faldini C, Ruffilli A. Identification of Epigenetic Biomarkers of Adolescent Idiopathic Scoliosis Progression: A Workflow to Assess Local Gene Expression. Int J Mol Sci 2024; 25:5329. [PMID: 38791368 PMCID: PMC11120692 DOI: 10.3390/ijms25105329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a three-dimensional structural deformity of the spine that affects 2-3% of adolescents under the age of 16. AIS etiopathogenesis is not completely understood; however, the disease phenotype is correlated to multiple genetic loci and results from genetic-environmental interactions. One of the primary, still unresolved issues is the implementation of reliable diagnostic and prognostic markers. For clinical management improvement, predictors of curve progression are particularly needed. Recently, an epigenetic contribution to AIS development and progression was proposed; nevertheless, validation of data obtained in peripheral tissues and identification of the specific mechanisms and genes under epigenetic control remain limited. In this study, we propose a methodological approach for the identification of epigenetic markers of AIS progression through an original workflow based on the preliminary characterization of local expression of candidate genes in tissues directly involved in the pathology. The feasibility of the proposed methodological protocol has been originally tested here in terms of identification of the putative epigenetic markers of AIS progression, collection of the different tissues, retrieval of an appropriate amount and quality of RNA and DNA, and identification of suitable reference genes.
Collapse
Affiliation(s)
- Simona Neri
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
| | - Elisa Assirelli
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
| | - Marco Manzetti
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Giovanni Viroli
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Marco Ialuna
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
| | - Matteo Traversari
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
| | - Jacopo Ciaffi
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
| | - Francesco Ursini
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (E.A.); (J.C.); (F.U.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Cesare Faldini
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| | - Alberto Ruffilli
- 1st Orthopaedic and Traumatologic Clinic, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.M.); (G.V.); (M.I.); (M.T.); (C.F.); (A.R.)
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
5
|
Lu W, Feng W, Lai J, Yuan D, Xiao W, Li Y. Role of adipokines in sarcopenia. Chin Med J (Engl) 2023; 136:1794-1804. [PMID: 37442757 PMCID: PMC10406092 DOI: 10.1097/cm9.0000000000002255] [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: 10/05/2022] [Indexed: 07/15/2023] Open
Abstract
ABSTRACT Sarcopenia is an age-related disease that mainly involves decreases in muscle mass, muscle strength and muscle function. At the same time, the body fat content increases with aging, especially the visceral fat content. Adipose tissue is an endocrine organ that secretes biologically active factors called adipokines, which act on local and distant tissues. Studies have revealed that some adipokines exert regulatory effects on muscle, such as higher serum leptin levels causing a decrease in muscle function and adiponectin inhibits the transcriptional activity of Forkhead box O3 (FoxO3) by activating peroxisome proliferators-activated receptor-γ coactivator -1α (PGC-1α) and sensitizing cells to insulin, thereby repressing atrophy-related genes (atrogin-1 and muscle RING finger 1 [MuRF1]) to prevent the loss of muscle mass. Here, we describe the effects on muscle of adipokines produced by adipose tissue, such as leptin, adiponectin, resistin, mucin and lipocalin-2, and discuss the importance of these adipokines for understanding the development of sarcopenia.
Collapse
Affiliation(s)
- Wenhao Lu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Wenjie Feng
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jieyu Lai
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Dongliang Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan 410083, China
| | - Wenfeng Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
6
|
Adiponectin Is a Contributing Factor of Low Appendicular Lean Mass in Older Community-Dwelling Women: A Cross-Sectional Study. J Clin Med 2022; 11:jcm11237175. [PMID: 36498747 PMCID: PMC9740541 DOI: 10.3390/jcm11237175] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Inflammation is a chronic, sterile, low-grade inflammation that develops with advanced age in the absence of overt infection and may contribute to the pathophysiology of sarcopenia, a progressive and generalized skeletal muscle disorder. Furthermore, a series of biomarkers linked to sarcopenia occurrence have emerged. To aid diagnostic and treatment strategies for low muscle mass in sarcopenia and other related conditions, the objective of this work was to investigate potential biomarkers associated with appendicular lean mass in community-dwelling older women. This is a cross-sectional study with 71 older women (75 ± 7 years). Dual-energy X-ray absorptiometry was used to assess body composition. Plasmatic blood levels of adipokines (i.e., adiponectin, leptin, and resistin), tumor necrosis factor (TNF) and soluble receptors (sTNFr1 and sTNFr2), interferon (INF), brain-derived neurotrophic factor (BDNF), and interleukins (IL-2, IL-4, IL-5, IL-6, IL-8, and IL-10) were determined by enzyme-linked immunosorbent assay. Older women with low muscle mass showed higher plasma levels of adiponectin, sTNFr1, and IL-8 compared to the regular muscle mass group. In addition, higher adiponectin plasma levels explained 14% of the lower appendicular lean mass. High adiponectin plasmatic blood levels can contribute to lower appendicular lean mass in older, community-dwelling women.
Collapse
|
7
|
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
|
8
|
Morinaga M, Sako N, Isobe M, Lee-Hotta S, Sugiura H, Kametaka S. Aerobic Exercise Ameliorates Cancer Cachexia-Induced Muscle Wasting through Adiponectin Signaling. Int J Mol Sci 2021; 22:3110. [PMID: 33803685 PMCID: PMC8002946 DOI: 10.3390/ijms22063110] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022] Open
Abstract
Cachexia is a multifactorial syndrome characterized by muscle loss that cannot be reversed by conventional nutritional support. To uncover the molecular basis underlying the onset of cancer cachectic muscle wasting and establish an effective intervention against muscle loss, we used a cancer cachectic mouse model and examined the effects of aerobic exercise. Aerobic exercise successfully suppressed muscle atrophy and activated adiponectin signaling. Next, a cellular model for cancer cachectic muscle atrophy using C2C12 myotubes was prepared by treating myotubes with a conditioned medium from a culture of colon-26 cancer cells. Treatment of the atrophic myotubes with recombinant adiponectin was protective against the thinning of cells through the increased production of p-mTOR and suppression of LC3-II. Altogether, these findings suggest that the activation of adiponectin signaling could be part of the molecular mechanisms by which aerobic exercise ameliorates cancer cachexia-induced muscle wasting.
Collapse
MESH Headings
- Adiponectin/genetics
- Adiponectin/metabolism
- Animals
- Cachexia/complications
- Cachexia/metabolism
- Cell Line, Tumor
- Culture Media, Conditioned/pharmacology
- Disease Models, Animal
- Female
- Mice, Inbred BALB C
- Microtubule-Associated Proteins/metabolism
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Muscular Atrophy/complications
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Phosphorylation/drug effects
- Physical Conditioning, Animal
- Protein Biosynthesis/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recombinant Proteins/pharmacology
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Makoto Morinaga
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, Aichi 461-0047, Japan
| | - Naoki Sako
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, Aichi 461-0047, Japan
| | - Mari Isobe
- Division of Morphological Sciences, Kagoshima University Graduate School of Medicine and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Sachiko Lee-Hotta
- Division of Creative Physical Therapy, Field of Prevention and Rehabilitation Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, Aichi 461-0047, Japan
| | - Hideshi Sugiura
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, Aichi 461-0047, Japan
| | - Satoshi Kametaka
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, Aichi 461-0047, Japan
| |
Collapse
|
9
|
Wilhelmsen A, Tsintzas K, Jones SW. Recent advances and future avenues in understanding the role of adipose tissue cross talk in mediating skeletal muscle mass and function with ageing. GeroScience 2021; 43:85-110. [PMID: 33528828 PMCID: PMC8050140 DOI: 10.1007/s11357-021-00322-4] [Citation(s) in RCA: 9] [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: 11/02/2020] [Accepted: 01/01/2021] [Indexed: 12/15/2022] Open
Abstract
Sarcopenia, broadly defined as the age-related decline in skeletal muscle mass, quality, and function, is associated with chronic low-grade inflammation and an increased likelihood of adverse health outcomes. The regulation of skeletal muscle mass with ageing is complex and necessitates a delicate balance between muscle protein synthesis and degradation. The secretion and transfer of cytokines, long non-coding RNAs (lncRNAs) and microRNAs (miRNAs), both discretely and within extracellular vesicles, have emerged as important communication channels between tissues. Some of these factors have been implicated in regulating skeletal muscle mass, function, and pathologies and may be perturbed by excessive adiposity. Indeed, adipose tissue participates in a broad spectrum of inter-organ communication and obesity promotes the accumulation of macrophages, cellular senescence, and the production and secretion of pro-inflammatory factors. Pertinently, age-related sarcopenia has been reported to be more prevalent in obesity; however, such effects are confounded by comorbidities and physical activity level. In this review, we provide evidence that adiposity may exacerbate age-related sarcopenia and outline some emerging concepts of adipose-skeletal muscle communication including the secretion and processing of novel myokines and adipokines and the role of extracellular vesicles in mediating inter-tissue cross talk via lncRNAs and miRNAs in the context of sarcopenia, ageing, and obesity. Further research using advances in proteomics, transcriptomics, and techniques to investigate extracellular vesicles, with an emphasis on translational, longitudinal human studies, is required to better understand the physiological significance of these factors, the impact of obesity upon them, and their potential as therapeutic targets in combating muscle wasting.
Collapse
Affiliation(s)
- Andrew Wilhelmsen
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Kostas Tsintzas
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK.
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Queen Elizabeth Hospital, The University of Birmingham, Birmingham, UK
| |
Collapse
|
10
|
Komici K, Dello Iacono A, De Luca A, Perrotta F, Bencivenga L, Rengo G, Rocca A, Guerra G. Adiponectin and Sarcopenia: A Systematic Review With Meta-Analysis. Front Endocrinol (Lausanne) 2021; 12:576619. [PMID: 33935962 PMCID: PMC8082154 DOI: 10.3389/fendo.2021.576619] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sarcopenia is a progressive loss of skeletal muscle mass whose pathophysiology has been proposed to possibly involve mechanisms of altered inflammatory status and endocrine function. Adiponectin has been shown to modulate inflammatory status and muscle metabolism. However, the possible association between adiponectin levels and sarcopenia is poorly understood. In order to fill this gap, in the present manuscript we aimed to summarize the current evidence with a systematic review and a meta-analysis of studies reporting serum adiponectin levels in patients with sarcopenia compared to non-sarcopenic controls. METHODS An electronic search through Medline/PubMed, Cochrane Library, and Science Direct was performed till March 1, 2020. From the included papers, meta-analysis of cross-sectional studies comparing serum levels of adiponectin between patients with sarcopenia and controls was performed. RESULTS Out of 1,370 initial studies, seven studies were meta-analyzed. Sarcopenic participants had significantly higher levels of adiponectin Hedges' g with 95% confidence interval (CI): 1.20 (0.19-2.22), p = 0.02 than controls. Subgroup analysis, performed in Asian population and focused on identification of the condition based on AWGS criteria, reported higher adiponectin levels in sarcopenic population (2.1 (0.17-4.03), p = 0.03 and I2 = 98.98%. Meta-regression analysis revealed female gender to significantly influence the results as demonstrated by beta = 0.14 (95% CI (0.010-0.280), p = 0.040). CONCLUSIONS Our meta-analysis found evidence that sarcopenia is associated with higher adiponectin levels. However, caution is warranted on the interpretation of these findings, and future longitudinal research is required to disentangle and better understand the topic.
Collapse
Affiliation(s)
- Klara Komici
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
- *Correspondence: Klara Komici,
| | - Antonio Dello Iacono
- School of Health and Life Sciences, University of the West of Scotland, Hamilton, United Kingdom
| | - Antonio De Luca
- Section of Human Anatomy, Department of Mental and Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Fabio Perrotta
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Leonardo Bencivenga
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Giuseppe Rengo
- Department of Translational Medical Sciences, University of Naples “Federico II”, Naples, Italy
- Istituti Clinici Scientifici Maugeri SpA Società Benefit (ICS Maugeri SpA SB), Telese Terme, Italy
| | - Aldo Rocca
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, Campobasso, Italy
| |
Collapse
|
11
|
Yu A, Zheng Y, Gong Y, Yu R, Yang Z, Chen X. Adiponectin promotes myogenic differentiation via a Mef2C-AdipoR1 positive feedback loop. Gene 2020; 771:145380. [PMID: 33359123 DOI: 10.1016/j.gene.2020.145380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 11/17/2020] [Accepted: 12/15/2020] [Indexed: 11/16/2022]
Abstract
Adiponectin is an important hormone that regulates systemic metabolism, and it has been reported that globular adiponectin promotes myogenic differentiation. However, the mechanisms by which adiponectin promotes myogenic differentiation is not fully understood. In the present study, we show that adiponectin and its receptor 1 are significantly up-regulated during myogenic differentiation and that adiponectin increased the expression level of a core myogenic regulator, Mef2C, which is required for the effects of adiponectin on promoting myogenic differentiation. A transcriptional inhibitor of Mef2C, HDAC9, was down-regulated by adiponectin. In turn, Mef2C overexpression up-regulates adiponectin and its receptor, AdipoR1, to increase myogenic differentiation. We showed that mechanistically, Mef2C directly binds to AdipoR1 promoter to transcriptionally up-regulate AdipoR1 expression, which is required for the effects of Mef2C overexpression on myogenic differentiation. Thus, adiponectin/AdipoR1 and Mef2c form a positive feedback loop to promote myogenic differentiation.
Collapse
Affiliation(s)
- An Yu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yu Zheng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Yuqing Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ruofan Yu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zaiqing Yang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China.
| |
Collapse
|
12
|
Abou-Samra M, Selvais CM, Dubuisson N, Brichard SM. Adiponectin and Its Mimics on Skeletal Muscle: Insulin Sensitizers, Fat Burners, Exercise Mimickers, Muscling Pills … or Everything Together? Int J Mol Sci 2020; 21:ijms21072620. [PMID: 32283840 PMCID: PMC7178193 DOI: 10.3390/ijms21072620] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/01/2020] [Accepted: 04/03/2020] [Indexed: 02/07/2023] Open
Abstract
Adiponectin (ApN) is a hormone abundantly secreted by adipocytes and it is known to be tightly linked to the metabolic syndrome. It promotes insulin-sensitizing, fat-burning, and anti-atherosclerotic actions, thereby effectively counteracting several metabolic disorders, including type 2 diabetes, obesity, and cardiovascular diseases. ApN is also known today to possess powerful anti-inflammatory/oxidative and pro-myogenic effects on skeletal muscles exposed to acute or chronic inflammation and injury, mainly through AdipoR1 (ApN specific muscle receptor) and AMP-activated protein kinase (AMPK) pathway, but also via T-cadherin. In this review, we will report all the beneficial and protective properties that ApN can exert, specifically on the skeletal muscle as a target tissue. We will highlight its effects and mechanisms of action, first in healthy skeletal muscle including exercised muscle, and second in diseased muscle from a variety of pathological conditions. In the end, we will go over some of AdipoRs agonists that can be easily produced and administered, and which can greatly mimic ApN. These interesting and newly identified molecules could pave the way towards future therapeutic approaches to potentially prevent or combat not only skeletal muscle disorders but also a plethora of other diseases with sterile inflammation or metabolic dysfunction.
Collapse
|
13
|
Abou-Samra M, Selvais CM, Boursereau R, Lecompte S, Noel L, Brichard SM. AdipoRon, a new therapeutic prospect for Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 2020; 11:518-533. [PMID: 31965757 PMCID: PMC7113498 DOI: 10.1002/jcsm.12531] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 10/23/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Adiponectin (ApN) is a hormone known to exhibit insulin-sensitizing, fat-burning, and anti-inflammatory properties in several tissues, including the skeletal muscle. Duchenne muscular dystrophy (DMD) is a devastating disease characterized by dystrophin deficiency with subsequent chronic inflammation, myofiber necrosis, and impaired regeneration. Previously, we showed that transgenic up-regulation of ApN could significantly attenuate the dystrophic phenotype in mdx mice (model of DMD). Recently, an orally active ApN receptor agonist, AdipoRon, has been identified. This synthetic small molecule has the advantage of being more easily produced and administrable than ApN. The aim of this study was to investigate the potential effects of AdipoRon on the dystrophic muscle. METHODS Four-week-old mdx mice (n = 6-9 per group) were orally treated with AdipoRon (mdx-AR) for 8 weeks and compared with untreated (mdx) mice and to control (wild-type) mice. In vivo functional tests were carried out to measure the global force and endurance of mice. Ex vivo biochemical and molecular analyses were performed to evaluate the pathophysiology of the skeletal muscle. Finally, in vitro tests were conducted on primary cultures of healthy and DMD human myotubes. RESULTS AdipoRon treatment mitigated oxidative stress (-30% to 45% for 4-hydroxy-2-nonenal and peroxiredoxin 3, P < 0.0001) as well as inflammation in muscles of mdx mice (-35% to 65% for interleukin 1 beta, tumour necrosis factor alpha, and cluster of differentiation 68, a macrophage maker, P < 0.0001) while increasing the anti-inflammatory cytokine, interleukin 10 (~5-fold, P < 0.0001). AdipoRon also improved the myogenic programme as assessed by a ~2-fold rise in markers of muscle proliferation and differentiation (P < 0.01 or less vs. untreated mdx). Plasma lactate dehydrogenase and creatine kinase were reduced by 30-40% in mdx-AR mice, reflecting less sarcolemmal damage (P < 0.0001). When compared with untreated mdx mice, mdx-AR mice exhibited enhanced physical performance with an increase in both muscle force and endurance and a striking restoration of the running capacity during eccentric exercise. AdipoRon mainly acted through ApN receptor 1 by increasing AMP-activated protein kinase signalling, which led to repression of nuclear factor-kappa B, up-regulation of utrophin (a dystrophin analogue), and a switch towards an oxidative and more resistant fibre phenotype. The effects of AdipoRon were then recapitulated in human DMD myotubes. CONCLUSIONS These results demonstrate that AdipoRon exerts several beneficial effects on the dystrophic muscle. This molecule could offer promising therapeutic prospect for managing DMD or other muscle and inflammatory disorders.
Collapse
Affiliation(s)
- Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Camille M Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Raphael Boursereau
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Sophie Lecompte
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| | - Sonia M Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
14
|
Mosele FC, Bissi Ricci R, Abreu P, Rosa Neto JC. Muscle regeneration in adiponectin knockout mice showed early activation of anti-inflammatory response with perturbations in myogenesis. J Cell Physiol 2020; 235:6183-6193. [PMID: 32003014 DOI: 10.1002/jcp.29547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Activation, proliferation, and differentiation of satellite cells can be influenced by extracellular factors, such as adiponectin. This adipokine has been proposed as a regulator of in vitro myogenesis, but its action on in vivo regeneration is not still elucidated. We used C57BL/6 (wild-type [WT]) and adiponectin knockout (AdKO) mice injured with barium chloride at periods of 3, 7, and 14 days after injury. The AdKO presented a higher number of centralized nuclei after 7 days, and a reduction in myogenic genes was observed after 3 days. Moreover, these mice presented an increase in anti-inflammatory cytokines after 3 and 7 days, and an increase in the M2 gene marker and proinflammatory cytokines after 7 days. The WT demonstrated an increase in adiponectin messenger RNA after 7 days. These results demonstrate that adiponectin is important in tissue remodeling during regeneration and that its deficiency does not compromise the maturation of muscle fibers, due to an increase in anti-inflammatory response; however, there is a possible impairment in proinflammatory response and an increase in centralized myonuclei.
Collapse
Affiliation(s)
- Francielle C Mosele
- Department of Cellular Biology and Development, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Rafael Bissi Ricci
- Department of Cellular Biology and Development, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Phablo Abreu
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, Brazil
| | - José C Rosa Neto
- Department of Cellular Biology and Development, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
15
|
Gamberi T, Magherini F, Mannelli M, Chrisam M, Cescon M, Castagnaro S, Modesti A, Braghetta P, Fiaschi T. Role of adiponectin in the metabolism of skeletal muscles in collagen VI-related myopathies. J Mol Med (Berl) 2019; 97:793-801. [PMID: 30927046 DOI: 10.1007/s00109-019-01766-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 02/11/2019] [Accepted: 03/05/2019] [Indexed: 12/31/2022]
Abstract
The role of adiponectin has been particularly deepened in diabetic muscles while the study of adiponectin in hereditary myopathies has been marginally investigated. Here, we report the study about adiponectin effects in Col6a1-/- (collagen VI-null) mice. Col6a1-/- mice show myophatic phenotype closer to that of patients with Bethlem myopathy, thus representing an excellent animal model for the study of this hereditary disease. Our findings demonstrate that Col6a1-/- mice have decreased plasma adiponectin content and diseased myoblasts have an impaired autocrine secretion of the hormone. Moreover, Col6a1-/- myoblasts show decreased glucose uptake and mitochondria with depolarized membrane potential and impaired functionality, as supported by decreased oxygen consumption. Exogenous addition of globular adiponectin modifies the features of Col6a1-/- myoblasts, becoming closer to that of the healthy myoblasts. Indeed, globular adiponectin enhances glucose uptake in Col6a1-/- myoblasts, modifies mitochondrial membrane potential, and restores oxygen consumption, turning closer to those of wild-type myoblasts. Finally, increase of plasma adiponectin level in Col6a1-/- mice is induced by fasting, a condition that has been previously shown to lead to the amelioration of the dystrophic phenotype. Collectively, our results demonstrate that exogenous replenishment of adiponectin reverses metabolic abnormalities observed in Col6a1-/- myoblasts. KEY MESSAGES: Col6a1-/- mice have decreased level of plasma adiponectin. Myoblasts from Col6a1-/- muscles have impaired local adiponectin secretion. Col6a1-/- myoblasts reveal altered metabolic features. Addition of exogenous adiponectin ameliorates Col6a1-/- metabolic features.
Collapse
Affiliation(s)
- Tania Gamberi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, viale Morgagni 50, 50134, Florence, Italy
| | - Francesca Magherini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, viale Morgagni 50, 50134, Florence, Italy
| | - Michele Mannelli
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, viale Morgagni 50, 50134, Florence, Italy
| | - Martina Chrisam
- Dipartimento di Medicina Molecolare, Università degli Studi di Padova, Padua, Italy
| | - Matilde Cescon
- Dipartimento di Medicina Molecolare, Università degli Studi di Padova, Padua, Italy
| | - Silvia Castagnaro
- Dipartimento di Medicina Molecolare, Università degli Studi di Padova, Padua, Italy
| | - Alessandra Modesti
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, viale Morgagni 50, 50134, Florence, Italy
| | - Paola Braghetta
- Dipartimento di Medicina Molecolare, Università degli Studi di Padova, Padua, Italy
| | - Tania Fiaschi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, viale Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
16
|
Adiponectin in Myopathies. Int J Mol Sci 2019; 20:ijms20071544. [PMID: 30934785 PMCID: PMC6480168 DOI: 10.3390/ijms20071544] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023] Open
Abstract
In skeletal muscle, adiponectin has varied and pleiotropic functions, ranging from metabolic, anti-inflammatory, insulin-sensitizing to regenerative roles. Despite the important functions exerted by adiponectin, the study of the hormone in myopathies is still marginal. Myopathies include inherited and non-inherited/acquired neuromuscular pathologies characterized by muscular degeneration and weakness. This review reports current knowledge about adiponectin in myopathies, regarding in particular the role of adiponectin in some hereditary myopathies (as Duchenne muscular dystrophy) and non-inherited/acquired myopathies (such as idiopathic inflammatory myopathies and fibromyalgia). These studies show that some myopathies are characterized by decreased concentration of plasma adiponectin and that hormone replenishment induces beneficial effects in the diseased muscles. Overall, these findings suggest that adiponectin could constitute a future new therapeutic approach for the improvement of the abnormalities caused by myopathies.
Collapse
|
17
|
Krause MP, Milne KJ, Hawke TJ. Adiponectin-Consideration for its Role in Skeletal Muscle Health. Int J Mol Sci 2019; 20:ijms20071528. [PMID: 30934678 PMCID: PMC6480271 DOI: 10.3390/ijms20071528] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 03/24/2019] [Accepted: 03/25/2019] [Indexed: 12/17/2022] Open
Abstract
Adiponectin regulates metabolism through blood glucose control and fatty acid oxidation, partly mediated by downstream effects of adiponectin signaling in skeletal muscle. More recently, skeletal muscle has been identified as a source of adiponectin expression, fueling interest in the role of adiponectin as both a circulating adipokine and a locally expressed paracrine/autocrine factor. In addition to being metabolically responsive, skeletal muscle functional capacity, calcium handling, growth and maintenance, regenerative capacity, and susceptibility to chronic inflammation are all strongly influenced by adiponectin stimulation. Furthermore, physical exercise has clear links to adiponectin expression and circulating concentrations in healthy and diseased populations. Greater physical activity is generally related to higher adiponectin expression while lower adiponectin levels are found in inactive obese, pre-diabetic, and diabetic populations. Exercise training typically restores plasma adiponectin and is associated with improved insulin sensitivity. Thus, the role of adiponectin signaling in skeletal muscle has expanded beyond that of a metabolic regulator to include several aspects of skeletal muscle function and maintenance critical to muscle health, many of which are responsive to, and mediated by, physical exercise.
Collapse
Affiliation(s)
- Matthew P Krause
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, 401 Sunset Avenue, Windsor, ON N9B 3P4, Canada.
| | - Kevin J Milne
- Department of Kinesiology, Faculty of Human Kinetics, University of Windsor, 401 Sunset Avenue, Windsor, ON N9B 3P4, Canada.
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, 1280 Main Street, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|
18
|
Zhou LN, Lin YN, Gu CJ, Zhou JP, Sun XW, Cai XT, Du J, Li QY. AMPK/FOXO1 signaling pathway is indispensable in visfatin-regulated myosin heavy chain expression in C2C12 myotubes. Life Sci 2019; 224:197-203. [PMID: 30926551 DOI: 10.1016/j.lfs.2019.03.060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 03/09/2019] [Accepted: 03/25/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Few studies have addressed the effects of visfatin on skeletal muscle remodeling. The aim of the study was to investigate the effects of visfatin on the expressions of myosin heavy chain (MHC) and its isoforms, the major indicator of fiber types and contractile properties of skeletal muscle. MATERIALS AND METHODS Levels of MHC, MHC I, MHC IIa, MHC IIb, adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), p-AMPK and forkhead box protein O1 (FOXO1) were tested in visfatin-treated C2C12 myotubes. C2C12 myotubes were treated with visfatin combined with AMPK inhibitor or AMPK activator to investigate the role of AMPK in visfatin-mediated MHC expression. FOXO1 was overexpressed or knocked down in C2C12 myotubes to explore the role of FOXO1 in visfatin-mediated MHC expression. RESULTS Compared with the vehicle group, treatment with 5 μg/ml visfatin increased the levels of total MHC and its isoforms, MHC I, MHC IIa and MHC IIb, by 1.93, 1.84, 1.80, and 1.92 folds, respectively (all p = 0,001). Visfatin suppressed AMPK phosphorylation and decreased FOXO1 expression in C2C12 myotubes. The effects of visfatin on MHC I and MHC IIa expression were canceled by AMPK activator AICAR. FOXO1 overexpression minimized the visfatin-induced upregulation of MHC I, MHC IIa and MHC IIb. The effect of AMPK activator AICAR on MHC and its isoforms expression was minimized by knockdown of FOXO1. CONCLUSIONS The findings revealed that visfatin promoted expressions of MHC and its isoforms in C2C12 myotubes via suppressing AMPK/FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Li Na Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Respiratory Medicine, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai 281000, China
| | - Ying Ni Lin
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chen Juan Gu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jian Ping Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xian Wen Sun
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao Ting Cai
- Department of Respiratory Medicine, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai 281000, China
| | - Juan Du
- Department of Respiratory Medicine, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai 281000, China
| | - Qing Yun Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
19
|
Liu Y, Vu V, Sweeney G. Examining the Potential of Developing and Implementing Use of Adiponectin-Targeted Therapeutics for Metabolic and Cardiovascular Diseases. Front Endocrinol (Lausanne) 2019; 10:842. [PMID: 31920962 PMCID: PMC6918867 DOI: 10.3389/fendo.2019.00842] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023] Open
Abstract
Cardiometabolic diseases encompass those affecting the heart and vasculature as well as other metabolic problems, such as insulin resistance, diabetes, and non-alcoholic fatty liver disease. These diseases tend to have common risk factors, one of which is impaired adiponectin action. This may be due to reduced bioavailability of the hormone or resistance to its effects on target tissues. A strong negative correlation between adiponectin levels and cardiometabolic diseases has been well-documented and research shown that adiponectin has cardioprotective, insulin sensitizing and direct beneficial metabolic effects. Thus, therapeutic approaches to enhance adiponectin action are widely considered to be desirable. The complexity of adiponectin structure and function has so far made progress in this area less than ideal. In this article we will review the effects and mechanism of action of adiponectin on cardiometabolic tissues, identify scenarios where enhancing adiponectin action would be of clinical value and finally discuss approaches via which this can be achieved.
Collapse
Affiliation(s)
- Ying Liu
- Metabolic Disease Research Division, iCarbonX Co. Ltd., Shenzhen, China
- *Correspondence: Ying Liu
| | - Vivian Vu
- Department of Biology, York University, Toronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
- Gary Sweeney
| |
Collapse
|
20
|
Activation of adiponectin receptors has negative impact on muscle mass in C2C12 myotubes and fast-type mouse skeletal muscle. PLoS One 2018; 13:e0205645. [PMID: 30308063 PMCID: PMC6181411 DOI: 10.1371/journal.pone.0205645] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/29/2018] [Indexed: 11/19/2022] Open
Abstract
This study investigated the effects of AdipoRon, which is an agonist for adiponectin receptor 1 (AdipoR1) and AdipoR2, on the protein content, myotube diameter, and number of nuclei per myotube of C2C12 cells and skeletal muscle mass in C57BL/6J mice. AdipoRon suppressed the protein content, myotube diameter, and number of nuclei per myotube of C2C12 cells of C2C12 myotubes in a dose-dependent manner. Adiponectin-associated decline of protein content, diameter, and number of nuclei per myotube in C2C12 myotubes was partially rescued by knockdown of AdipoR1 and/or AdipoR2. Phosphorylation level of AMPK showed a trend to be increased by AdipoRon. A significant increase in phosphorylation level of AMPK was observed at 20 μM AdipoRon. Knockdown of AdipoR1 and/or AdipoR2 rescued AdipoRon-associated decrease in protein content of C2C12 myotubes. AdipoRon-associated increase in phosphorylation level of AMPK in C2C12 myotubes was suppressed by knockdown of AdipoR1 and/or AdipoR2. Successive intravenous injections of AdipoRon into mice caused a decrease in the wet weight of plantaris muscle (PLA), but not in soleus muscle (SOL). Mean fiber cross-sectional area of PLA, but not of SOL, was significantly decreased by AdipoRon administration. On the one hand, the expression level of phosphorylated AMPK and ubiquitinated protein in SOL and PLA muscles was upregulated by AdipoRon administration. On the other hand, AdipoRon administration induced no changes in the expression level of puromycin-labeled proteins in both SOL and PLA muscles. Expression level of adiponectin in extensor digitorum longus (EDL) muscle was increased by aging, but not in SOL muscle. Aging had no effect on the expression level of AdipoR1 and AdipoR2 in both muscles. Phosphorylation level of AMPK in EDL was increased by aging, but not SOL muscle. Results from this study suggest that high level of circulating adiponectin may induce skeletal muscle atrophy, especially fast-type muscle.
Collapse
|
21
|
Jiang H, Yang F, Lin T, Shao W, Meng Y, Ma J, Wang C, Gao R, Zhou X. Asymmetric expression of H19 and ADIPOQ in concave/convex paravertebral muscles is associated with severe adolescent idiopathic scoliosis. Mol Med 2018; 24:48. [PMID: 30241458 PMCID: PMC6145194 DOI: 10.1186/s10020-018-0049-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 09/10/2018] [Indexed: 12/19/2022] Open
Abstract
Background Adolescent idiopathic scoliosis (AIS) is the most common paediatric spinal deformity. The etiology and pathology of AIS remain unexplained, and have been reported to involve a combination of genetic and epigenetic factors. Since paravertebral muscle imbalance plays an important role in the onset and progression of scoliosis, we aimed to investigate transcriptomic differences by RNA-seq and identify significantly differentially expressed transcripts in two sides of paravertebral muscle in AIS. Methods RNA-seq was performed on 5 pairs of paravertebral muscle from 5 AIS patients. Significantly differentially expressed transcripts were validated by quantitative reverse polymerase chain reaction. Gene expression difference was correlated to clinical characteristics. Results We demonstrated that ADIPOQ mRNA and H19 is significantly differentially expressed between two sides of paravertebral muscle, relatively specific in the context of AIS. Relatively low H19 and high ADIPOQ mRNA expression levels in concave-sided muscle are associated with larger spinal curve and earlier age at initiation. We identified miR-675-5p encoded by H19 as a mechanistic regulator of ADIPOQ expression in AIS. We demonstrated that significantly reduced CCCTC-binding factor (CCTF) occupancy in the imprinting control region (ICR) of the H19 gene in the concave-sided muscle contributes to down-regulated H19 expression. Conclusions RNA-seq revealed transcriptomic differences between two sides of paravertebral muscle in AIS patients. Our findings imply that transcriptomic differences caused by epigenetic factors in affected individuals may account for the structural and functional imbalance of paravertebral muscle, which can expand our etiologic understanding of this disease. Electronic supplementary material The online version of this article (10.1186/s10020-018-0049-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China
| | - Fu Yang
- Department of Medical Genetics, Second Military Medical University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Cell Engineering (14DZ2272300), Shanghai, People's Republic of China
| | - Tao Lin
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China
| | - Wei Shao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China
| | - Yichen Meng
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China
| | - Jun Ma
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China
| | - Ce Wang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China
| | - Rui Gao
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China.
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, No.415 Fengyang Road, Shanghai, People's Republic of China.
| |
Collapse
|
22
|
Pal China S, Sanyal S, Chattopadhyay N. Adiponectin signaling and its role in bone metabolism. Cytokine 2018; 112:116-131. [PMID: 29937410 DOI: 10.1016/j.cyto.2018.06.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/06/2018] [Accepted: 06/09/2018] [Indexed: 12/14/2022]
Abstract
Adiponectin, the most prevalent adipo-cytokine in plasma plays critical metabolic and anti-inflammatory roles is fast emerging as an important molecular target for the treatment of metabolic disorders. Adiponectin action is critical in multiple organs including cardio-vascular system, muscle, liver, adipose tissue, brain and bone. Adiponectin signaling in bone has been a topic of active investigation lately. Human association studies and multiple mice models of gene deletion/modification failed to define a clear cause and effect of adiponectin signaling in bone. The most plausible reason could be the multimeric forms of adiponectin that display differential binding to receptors (adipoR1 and adipoR2) with cell-specific receptor variants in bone. Discovery of small molecule agonist of adipoR1 suggested a salutary role of this receptor in bone metabolism. The downstream signaling of adipoR1 in osteoblasts involves stimulation of oxidative phosphorylation leading to increased differentiation via the likely suppression of wnt inhibitor, sclerostin. On the other hand, the inflammation modulatory effect of adiponectin signaling suppresses the RANKL (receptor activator of nuclear factor κ-B ligand) - to - OPG (osteprotegerin) ratio in osteoblasts leading to the suppression of osteoclastogenic response. This review will discuss the adiponectin signaling and its role in skeletal homeostasis and critically assess whether adipoR1 could be a therapeutic target for the treatment of metabolic bone diseases.
Collapse
Affiliation(s)
- Shyamsundar Pal China
- Division of Endocrinology and CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226 031, India
| | - Sabyasachi Sanyal
- Division of Biochemistry, CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226 031, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and CSIR-Central Drug Research Institute, Sitapur Road, Lucknow 226 031, India.
| |
Collapse
|
23
|
Gamberi T, Magherini F, Modesti A, Fiaschi T. Adiponectin Signaling Pathways in Liver Diseases. Biomedicines 2018; 6:biomedicines6020052. [PMID: 29735928 PMCID: PMC6027295 DOI: 10.3390/biomedicines6020052] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 02/07/2023] Open
Abstract
In the liver, adiponectin regulates both glucose and lipid metabolism and exerts an insulin-sensitizing effect. The binding of adiponectin with its specific receptors induces the activation of a proper signaling cascade that becomes altered in liver pathologies. This review describes the different signaling pathways in healthy and diseased hepatocytes, also highlighting the beneficial role of adiponectin in autophagy activation and hepatic regeneration.
Collapse
Affiliation(s)
- Tania Gamberi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Francesca Magherini
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Alessandra Modesti
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| | - Tania Fiaschi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche "Mario Serio", Università degli Studi di Firenze, Viale Morgagni 50, 50134 Firenze, Italy.
| |
Collapse
|
24
|
Wang L, Xue K, Wang Y, Niu L, Li L, Zhong T, Guo J, Feng J, Song T, Zhang H. Molecular and functional characterization of the adiponectin (AdipoQ) gene in goat skeletal muscle satellite cells. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2018; 31:1088-1097. [PMID: 29381891 PMCID: PMC6043445 DOI: 10.5713/ajas.17.0407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 01/30/2018] [Indexed: 01/21/2023]
Abstract
Objective It is commonly accepted that adiponectin binds to its two receptors to regulate fatty acid metabolism in adipocytes. To better understand their functions in the regulation of intramuscular adipogenesis in goats, we cloned the three genes (adiponectin [AdipoQ], adiponectin receptor 1 [AdipoR1], and AdipoR2) encoding these proteins and detected their mRNA distribution in different tissues. We also determined the role of AdipoQ in the adipogenic differentiation of goat skeletal muscle satellite cells (SMSCs). Methods SMSCs were isolated using 1 mg/mL Pronase E from the longissimus dorsi muscles of 3-day-old female Nanjiang brown goats. Adipogenic differentiation was induced in satellite cells by transferring the cells to Dulbecco’s modified Eagle’s medium supplemented with an isobutylmethylxanthine, dexamethasone and insulin cocktail. The pEGFP-N1-AD plasmid was transfected into SMSCs using Lipofectamine 2000. Expression of adiponectin in tissues and SMSCs was detected by quantitative polymerase chain reaction and immunocytochemical staining. Results The three genes were predominantly expressed in adipose and skeletal muscle tissues. According to fluorescence and immunocytochemical analyses, adiponectin protein expression was only observed in the cytoplasm, suggesting that adiponectin is localized to the cytoplasm of goat SMSCs. In SMSCs overexpressing the AdipoQ gene, adiponectin promoted SMSC differentiation into adipocytes and significantly (p<0.05) up-regulated expression of AdipoR2, acetyl-CoA carboxylase, fatty-acid synthase, and sterol regulatory element-binding protein-1, though expression of CCAAT/enhancer-binding protein-α, peroxisome proliferator-activated receptor γ, and AdipoR1 did not change significantly. Conclusion Adiponectin induced SMSC differentiation into adipocytes, indicating that adiponectin may promote intramuscular adipogenesis in goat SMSC.
Collapse
Affiliation(s)
- Linjie Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ke Xue
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lili Niu
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Li Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Tao Zhong
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jiazhong Guo
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jing Feng
- Institute of Animal Science, Tibet Academy of Agricultural & Animal Husbandry Science, Lhasa, China
| | - Tianzeng Song
- Institute of Animal Science, Tibet Academy of Agricultural & Animal Husbandry Science, Lhasa, China
| | - Hongping Zhang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Sun Q, Qi W, Xiao X, Yang SH, Kim D, Yoon KS, Clark JM, Park Y. Imidacloprid Promotes High Fat Diet-Induced Adiposity in Female C57BL/6J Mice and Enhances Adipogenesis in 3T3-L1 Adipocytes via the AMPKα-Mediated Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:6572-6581. [PMID: 28704996 PMCID: PMC5576855 DOI: 10.1021/acs.jafc.7b02584] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Imidacloprid, a neonicotinoid insecticide, was previously reported to enhance adipogenesis and resulted in insulin resistance in cell culture models. It was also reported to promote high fat diet-induced obesity and insulin resistance in male C57BL/6J mice. Thus, the goal of the present study was to determine the effects of imidacloprid and dietary fat interaction on the development of adiposity and insulin resistance in female C57BL/6J mice. Mice were fed with a low (4% w/w) or high fat (20% w/w) diet containing imidacloprid (0.06, 0.6, or 6 mg/kg bw/day) for 12 weeks. Mice fed with imidacloprid (0.6 mg/kg bw/day) significantly enhanced high fat diet-induced weight gain and adiposity. Treatment with imidacloprid significantly increased serum insulin levels with high fat diet without effects on other markers of glucose homeostasis. AMPKα activation was significantly inhibited by 0.6 and 6 mg imidacloprid/kg bw/day in white adipose tissue. Moreover, AMPKα activation with 5-aminoimidazole-4-carboxamide ribonucleotide abolished the effects of imidacloprid (10 μM) on enhanced adipogenesis in 3T3-L1 adipocytes. N-Acetyl cysteine also partially reversed the effects of imidacloprid on reduced phosphorylation of protein kinase B (AKT) in C2C12 myotubes. These results indicate that imidacloprid may potentiate high fat diet-induced adiposity in female C57BL/6J mice and enhance adipogenesis in 3T3-L1 adipocytes via the AMPKα-mediated pathway. Imidacloprid might also influence glucose homeostasis partially by inducing cellular oxidative stress in C2C12 myotubes.
Collapse
Affiliation(s)
- Quancai Sun
- Department
of Food Science, Department of Mathematics and Statistics, and Department of Veterinary and Animal
Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Weipeng Qi
- Department
of Food Science, Department of Mathematics and Statistics, and Department of Veterinary and Animal
Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Xiao Xiao
- Department
of Food Science, Department of Mathematics and Statistics, and Department of Veterinary and Animal
Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Szu-Hao Yang
- Department
of Food Science, Department of Mathematics and Statistics, and Department of Veterinary and Animal
Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Daeyoung Kim
- Department
of Food Science, Department of Mathematics and Statistics, and Department of Veterinary and Animal
Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Kyong Sup Yoon
- Department
of Biological Sciences and Environmental Sciences Program, Southern Illinois University, Edwardsville, Illinois 62026, United States
| | - John M. Clark
- Department
of Food Science, Department of Mathematics and Statistics, and Department of Veterinary and Animal
Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Yeonhwa Park
- Department
of Food Science, Department of Mathematics and Statistics, and Department of Veterinary and Animal
Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Tel: (413) 545-1018; e-mail:
| |
Collapse
|
26
|
Harada H, Kai H, Shibata R, Niiyama H, Nishiyama Y, Murohara T, Yoshida N, Katoh A, Ikeda H. New diagnostic index for sarcopenia in patients with cardiovascular diseases. PLoS One 2017; 12:e0178123. [PMID: 28542531 PMCID: PMC5436875 DOI: 10.1371/journal.pone.0178123] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/07/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Sarcopenia is an aging and disease-related syndrome characterized by progressive and generalized loss of skeletal muscle mass and strength, with the risk of frailty and poor quality of life. Sarcopenia is diagnosed by a decrease in skeletal muscle index (SMI) and reduction of either handgrip strength or gait speed. However, measurement of SMI is difficult for general physicians because it requires special equipment for bioelectrical impedance assay or dual-energy X-ray absorptiometry. The purpose of this study was, therefore, to explore a novel, simple diagnostic method of sarcopenia evaluation in patients with cardiovascular diseases (CVD). METHODS We retrospectively investigated 132 inpatients with CVD (age: 72±12 years, age range: 27-93 years, males: 61%) Binomial logistic regression and correlation analyses were used to assess the associations of sarcopenia with simple physical data and biomarkers, including muscle-related inflammation makers and nutritional markers. RESULTS Sarcopenia was present in 29.5% of the study population. Serum concentrations of adiponectin and sialic acid were significantly higher in sarcopenic than non-sarcopenic CVD patients. Stepwise multivariate binomial logistic regression analysis revealed that adiponectin, sialic acid, sex, age, and body mass index were independent factors for sarcopenia detection. Sarcopenia index, obtained from the diagnostic regression formula for sarcopenia detection including the five independent factors, indicated a high accuracy in ROC curve analysis (sensitivity 94.9%, specificity 69.9%) and the cutoff value for sarcopenia detection was -1.6134. Sarcopenia index had a significant correlation with the conventional diagnostic parameters of sarcopenia. CONCLUSIONS Our new sarcopenia index using simple parameters would be useful for diagnosing sarcopenia in CVD patients.
Collapse
Affiliation(s)
- Haruhito Harada
- Department of Cardiology, Kurume University Medical Center, Kurume, Japan
- * E-mail:
| | - Hisashi Kai
- Department of Cardiology, Kurume University Medical Center, Kurume, Japan
| | - Rei Shibata
- Department of Advanced Cardiovascular Therapeutics, Nagoya University, Nagoya, Japan
| | - Hiroshi Niiyama
- Department of Cardiology, Kurume University Medical Center, Kurume, Japan
| | - Yasuhiro Nishiyama
- Department of Cardiology, Kurume University Medical Center, Kurume, Japan
| | | | - Noriko Yoshida
- Institute of Health and Sports Sciences, Kurume University, Kurume, Japan
| | - Atsushi Katoh
- Department of Cardiology, Kurume University Medical Center, Kurume, Japan
| | - Hisao Ikeda
- Department of Physical Therapy, Faculty of Fukuoka Medical Technology, Teikyo University, Omuta, Japan
| |
Collapse
|
27
|
Abou-Samra M, Boursereau R, Lecompte S, Noel L, Brichard SM. Potential Therapeutic Action of Adiponectin in Duchenne Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1577-1585. [PMID: 28463682 DOI: 10.1016/j.ajpath.2017.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 02/27/2017] [Indexed: 01/11/2023]
Abstract
Adiponectin (ApN) is a hormone that exhibits anti-inflammatory effects on skeletal muscle exposed to acute and chronic inflammation. We have previously tested the implication of ApN in Duchenne muscular dystrophy (DMD) using mdx mice, a model of DMD, and by generating transgenic mdx mice overexpressing ApN. We showed that ApN can act as a preventive agent and delay disease progression by reducing muscle inflammation/injury and improving force/myogenesis. Herein, we took an opposite approach and crossed mdx mice with ApN knockout mice, to obtain mdx mice with ApN depletion. The aims were to test whether ApN deficiency could worsen the mdx phenotype and whether ApN supplementation can reverse several muscle abnormalities once the disease is settled. mdx-knockout mice exhibited lower muscle force/endurance as well as increased muscle damage when compared to regular mdx mice. Local administration of the ApN gene significantly reduced the expression of several oxidative stress/inflammatory markers and increased the expression of myogenic markers in the skeletal muscle. Finally, the presence of ApN markedly reduced the activity of NF-κB, a key player in muscle inflammation and myogenesis. ApN proves to be a powerful protector of the skeletal muscle capable of reversing the disease progression, thus making it a potential therapeutic agent for DMD.
Collapse
Affiliation(s)
- Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, Brussels, Belgium
| | - Raphaël Boursereau
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, Brussels, Belgium
| | - Sophie Lecompte
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, Brussels, Belgium
| | - Sonia M Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, Brussels, Belgium.
| |
Collapse
|
28
|
Skeletal muscle secretome in Duchenne muscular dystrophy: a pivotal anti-inflammatory role of adiponectin. Cell Mol Life Sci 2017; 74:2487-2501. [PMID: 28188344 PMCID: PMC5487898 DOI: 10.1007/s00018-017-2465-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 12/19/2016] [Accepted: 01/11/2017] [Indexed: 12/20/2022]
Abstract
Background Persistent inflammation exacerbates the progression of Duchenne muscular dystrophy (DMD). The hormone, adiponectin (ApN), which is decreased in the metabolic syndrome, exhibits anti-inflammatory properties on skeletal muscle and alleviates the dystrophic phenotype of mdx mice. Here, we investigate whether ApN retains its anti-inflammatory action in myotubes obtained from DMD patients. We unravel the underlying mechanisms by studying the secretome and the early events of ApN. Methods Primary cultures of myotubes from DMD and control patients were treated or not by ApN after an inflammatory challenge. Myokines secreted in medium were identified by cytokine antibody-arrays and ELISAs. The early events of ApN signaling were assessed by abrogating selected genes. Results ApN retained its anti-inflammatory properties in both dystrophic and control myotubes. Profiling of secretory products revealed that ApN downregulated the secretion of two pro-inflammatory factors (TNFα and IL-17A), one soluble receptor (sTNFRII), and one chemokine (CCL28) in DMD myotubes, while upregulating IL-6 that exerts some anti-inflammatory effects. These changes were explained by pretranslational mechanisms. Earlier events of the ApN cascade involved AdipoR1, the main receptor for muscle, and the AMPK-SIRT1-PGC-1α axis leading, besides alteration of the myokine profile, to the upregulation of utrophin A (a dystrophin analog). Conclusion ApN retains its beneficial properties in dystrophic muscles by activating the AdipoR1-AMPK-SIRT1-PGC-1α pathway, thereby inducing a shift in the secretion of downstream myokines toward a less inflammatory profile while upregulating utrophin. ApN, the early events of the cascade and downstream myokines may be therapeutic targets for the management of DMD.
Collapse
|
29
|
Singh AK, Shree S, Chattopadhyay S, Kumar S, Gurjar A, Kushwaha S, Kumar H, Trivedi AK, Chattopadhyay N, Maurya R, Ramachandran R, Sanyal S. Small molecule adiponectin receptor agonist GTDF protects against skeletal muscle atrophy. Mol Cell Endocrinol 2017; 439:273-285. [PMID: 27645900 DOI: 10.1016/j.mce.2016.09.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/23/2016] [Accepted: 09/14/2016] [Indexed: 02/02/2023]
Abstract
Skeletal muscle atrophy is a debilitating response to several major diseases, muscle disuse and chronic steroid treatment for which currently no therapy is available. Since adiponectin signaling plays key roles in muscle energetics, we assessed if globular adiponectin (gAd) or the small molecule adiponectin mimetic 6-C-β-D-glucopyranosyl-(2S,3S)-(+)-5,7,3',4'-tetrahydroxydihydroflavonol (GTDF) could ameliorate muscle atrophy. Both GTDF and gAd induced C2C12 myoblast differentiation. GTDF and gAd effectively prevented reduction in myotube area and suppressed the expressions of atrophy markers; atrogin-1 and muscle ring finger protein-1 (MuRF1) in models of steroid, cytokine and starvation -induced muscle atrophy. The protective effects of GTDF and gAd were routed through AMPK and AKT activation and thereby stimulation of PPAR gamma coactivator 1α and inhibition of forkhead box O transcription factors. Finally, GTDF and gAd mitigated dexamethasone-induced muscle atrophy in vivo. Together, our results demonstrate that activating adiponectin signaling may be an effective therapeutic strategy against skeletal muscle atrophy.
Collapse
Affiliation(s)
- Abhishek Kumar Singh
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Sonal Shree
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Sourav Chattopadhyay
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), CSIR- Central Drug Research Institute Campus, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Sudhir Kumar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Anagha Gurjar
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), CSIR- Central Drug Research Institute Campus, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Sapana Kushwaha
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Harish Kumar
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Arun Kumar Trivedi
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Rakesh Maurya
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Ravishankar Ramachandran
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India
| | - Sabyasachi Sanyal
- Division of Biochemistry, CSIR-Central Drug Research Institute, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India; Academy of Scientific and Innovative Research (AcSIR), CSIR- Central Drug Research Institute Campus, 10, Janakipuram Extn, Sitapur Road, Lucknow 226031, India.
| |
Collapse
|
30
|
Gamberi T, Modesti A, Magherini F, D'Souza DM, Hawke T, Fiaschi T. Activation of autophagy by globular adiponectin is required for muscle differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:694-702. [DOI: 10.1016/j.bbamcr.2016.01.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 01/14/2023]
|
31
|
Sente T, Van Berendoncks AM, Fransen E, Vrints CJ, Hoymans VY. Tumor necrosis factor-α impairs adiponectin signalling, mitochondrial biogenesis, and myogenesis in primary human myotubes cultures. Am J Physiol Heart Circ Physiol 2016; 310:H1164-75. [PMID: 26921438 DOI: 10.1152/ajpheart.00831.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/12/2016] [Indexed: 02/06/2023]
Abstract
Skeletal muscle metabolic changes are common in patients with chronic heart failure (HF). Previously, we demonstrated a functional skeletal muscle adiponectin resistance in HF patients with reduced left ventricular ejection fraction (HFrEF). We aimed to examine the impact of adiponectin receptor 1 (AdipoR1) deficiency and TNF-α treatment on adiponectin signaling, proliferative capacity, myogenic differentiation, and mitochondrial biogenesis in primary human skeletal muscle cells. Primary cultures of myoblasts and myotubes were initiated from the musculus vastus lateralis of 10 HFrEF patients (left ventricular ejection fraction; 31.30 ± 2.89%) and 10 age- and gender-matched healthy controls. Healthy control cultures were transfected with siAdipoR1 and/or exposed to TNF-α (10 ng/ml; 72 h). Primary cultures from HFrEF patients preserved the features of adiponectin resistance in vivo. AdipoR1 mRNA was negatively correlated with time to reach maximal cell index (r = -0.7319, P = 0.003). SiRNA-mediated AdipoR1 silencing reduced pAMPK (P < 0.01), AMPK activation (P = 0.046), and myoblast proliferation rate (xCELLigence Real-Time Cellular Analysis; P < 0.0001). Moreover, TNF-α decreased the mRNA expression of genes involved in glucose (APPL1, P = 0.0002; AMPK, P = 0.021), lipid (PPARα, P = 0.025; ACADM, P = 0.003), and mitochondrial (FOXO3, P = 0.018) metabolism, impaired myogenesis (MyoD1, P = 0.053; myogenin, P = 0.048) and polarized cytokine secretion toward a growth-promoting phenotype (IL-10, IL-1β, IFN-γ, P < 0.05 for all; Meso Scale Discovery Technology). Major features of adiponectin resistance are retained in primary cultures from the skeletal muscle of HFrEF patients. In addition, our results suggest that an increased inflammatory constitution contributes to adiponectin resistance and confers alterations in skeletal muscle differentiation, growth, and function.
Collapse
Affiliation(s)
- Tahnee Sente
- Laboratory for Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Wilrijk, Belgium; and
| | - An M Van Berendoncks
- Laboratory for Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Wilrijk, Belgium; and
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, Antwerp, Belgium
| | - Christiaan J Vrints
- Laboratory for Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Wilrijk, Belgium; and
| | - Vicky Y Hoymans
- Laboratory for Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Wilrijk, Belgium; and
| |
Collapse
|
32
|
Marique L, Senou M, Craps J, Delaigle A, Van Regemorter E, Wérion A, Van Regemorter V, Mourad M, Nyssen-Behets C, Lengelé B, Baldeschi L, Boschi A, Brichard S, Daumerie C, Many MC. Oxidative Stress and Upregulation of Antioxidant Proteins, Including Adiponectin, in Extraocular Muscular Cells, Orbital Adipocytes, and Thyrocytes in Graves' Disease Associated with Orbitopathy. Thyroid 2015; 25:1033-42. [PMID: 26176182 DOI: 10.1089/thy.2015.0087] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND Graves' orbitopathy (GO) is the main extrathyroidal manifestation associated with Graves' disease (GD). It is characterized by reduced eye motility due to an increased volume of orbital fat and/or of extraocular muscles (EOMs) infiltrated by fibrosis and adipose tissue. The pathogenetic mechanisms leading to fibrosis and adipogenesis are mainly based on the interaction between orbital fibroblasts and immune cells (lymphocytes and mast cells) infiltrating the GO EOMs. METHODS Analysis of the morphological status, oxidative stress (OS), and antioxidant defenses in the orbital muscular cells and adipocytes in GO patients compared with controls was conducted. RESULTS Both cell types are affected by OS, as shown by the increased expression of 4-hydroxynonenal, which leads to apoptosis in muscular cells. However, the EOMs and the adipocytes possess antioxidant defenses (peroxiredoxin 5 and catalase) against the OS, which are also upregulated in thyrocytes in GD. The expression of adiponectin (ApN) and proliferator-activated receptor gamma (PPARγ) is also increased in GO muscular cells and adipocytes. OS and antioxidant proteins expression are correlated to the level of blood antithyrotropin receptor antibodies (TSHR-Ab). CONCLUSION Even when TSHR-Ab level is normalized, OS and antioxidant protein expression is high in EOM muscular cells and adipocytes in GO compared with controls. This justifies a supplementation with antioxidants in active as well as chronic GO patients. Orbital muscular cells are also the sources of PPARγ and ApN, which have direct or indirect local protective effects against OS. Modulation of these proteins could be considered as a future therapeutic approach for GO.
Collapse
Affiliation(s)
- Lancelot Marique
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Maximin Senou
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Julie Craps
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Aurélie Delaigle
- 2 Pôles d'Endocrinologie, Diabète, et Nutrition, Institut de recherche Expérimentale et Clinique, Université Catholique de Louvain , Brussels, Belgium
| | | | - Alexis Wérion
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | | | - Michel Mourad
- 3 Département de Chirurgie Endocrinienne et de Trasplantation rénale, Université Catholique de Louvain , Brussels, Belgium
| | | | - Benoit Lengelé
- 1 Pôles de Morphologie, Université Catholique de Louvain , Brussels, Belgium
| | - Lelio Baldeschi
- 4 Département d'Ophtalmologie, Université Catholique de Louvain , Brussels, Belgium
| | - Antonella Boschi
- 4 Département d'Ophtalmologie, Université Catholique de Louvain , Brussels, Belgium
| | - Sonia Brichard
- 2 Pôles d'Endocrinologie, Diabète, et Nutrition, Institut de recherche Expérimentale et Clinique, Université Catholique de Louvain , Brussels, Belgium
- 5 Département d'Endocrinologie, Secteur des Sciences de la santé, Faculté de Médecine et de Médecine Dentaire, Université Catholique de Louvain , Brussels, Belgium
| | - Chantal Daumerie
- 5 Département d'Endocrinologie, Secteur des Sciences de la santé, Faculté de Médecine et de Médecine Dentaire, Université Catholique de Louvain , Brussels, Belgium
| | | |
Collapse
|
33
|
A novel adipokine C1q/TNF-related protein 3 is expressed in developing skeletal muscle and controls myoblast proliferation and differentiation. Mol Cell Biochem 2015; 409:271-82. [DOI: 10.1007/s11010-015-2531-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 08/06/2015] [Indexed: 01/08/2023]
|
34
|
Abou-Samra M, Lecompte S, Schakman O, Noel L, Many MC, Gailly P, Brichard SM. Involvement of adiponectin in the pathogenesis of dystrophinopathy. Skelet Muscle 2015; 5:25. [PMID: 26257862 PMCID: PMC4528853 DOI: 10.1186/s13395-015-0051-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/22/2015] [Indexed: 12/23/2022] Open
Abstract
Background The hormone adiponectin (ApN) is decreased in the metabolic syndrome, where it plays a key pathogenic role. ApN also exerts some anti-inflammatory effects on skeletal muscles in mice exposed to acute or chronic inflammation. Here, we investigate whether ApN could be sufficiently potent to counteract a severe degenerative muscle disease, with an inflammatory component such as Duchenne muscular dystrophy (DMD). Methods Mdx mice (a DMD model caused by dystrophin mutation) were crossed with mice overexpressing ApN in order to generate mdx-ApN mice; only littermates were used. Different markers of inflammation/oxidative stress and components of signaling pathways were studied. Global force was assessed by in vivo functional tests, and muscle injury with Evans Blue Dye (EBD). Eventually, primary cultures of human myotubes were used. Results Circulating ApN was markedly diminished in mdx mice. Replenishment of ApN strikingly reduced muscle inflammation, oxidative stress, and enhanced the expression of myogenic differentiation markers along with that of utrophin A (a dystrophin analog) in mdx-ApN mice. Accordingly, mdx-ApN mice exhibited higher global force and endurance as well as decreased muscle damage as quantified by curtailed extravasation of EBD in myofibers. These beneficial effects of ApN were recapitulated in human myotubes. ApN mediates its protection via the adiponectin receptor 1 (AdipoR1, the main ApN receptor in muscle) and the AMPK-SIRT1-PGC-1α signaling pathway, leading to downregulation of the nuclear factor kappa B (NF-κB) and inflammatory genes, together with upregulation of utrophin. Conclusions Adiponectin proves to be an extremely powerful hormone capable of protecting the skeletal muscle against inflammation and injury, thereby offering novel therapeutic perspectives for dystrophinopathies. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0051-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Sophie Lecompte
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Olivier Schakman
- Cellular and Molecular Unit, Institute of Neuroscience, Medical Sector, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Marie C Many
- Experimental Morphology Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Philippe Gailly
- Cellular and Molecular Unit, Institute of Neuroscience, Medical Sector, Catholic University of Louvain, 1200 Brussels, Belgium
| | - Sonia M Brichard
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Catholic University of Louvain, 1200 Brussels, Belgium
| |
Collapse
|
35
|
Laws KM, Sampson LL, Drummond-Barbosa D. Insulin-independent role of adiponectin receptor signaling in Drosophila germline stem cell maintenance. Dev Biol 2015; 399:226-36. [PMID: 25576925 DOI: 10.1016/j.ydbio.2014.12.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/02/2014] [Accepted: 12/29/2014] [Indexed: 10/24/2022]
Abstract
Adipocytes have key endocrine roles, mediated in large part by secreted protein hormones termed adipokines. The adipokine adiponectin is well known for its role in sensitizing peripheral tissues to insulin, and several lines of evidence suggest that adiponectin might also modulate stem cells/precursors. It remains unclear, however, how adiponectin signaling controls stem cells and whether this role is secondary to its insulin-sensitizing effects or distinct. Drosophila adipocytes also function as an endocrine organ and, although no obvious adiponectin homolog has been identified, Drosophila AdipoR encodes a well-conserved homolog of mammalian adiponectin receptors. Here, we generate a null AdipoR allele and use clonal analysis to demonstrate an intrinsic requirement for AdipoR in germline stem cell (GSC) maintenance in the Drosophila ovary. AdipoR null GSCs are not fully responsive to bone morphogenetic protein ligands from the niche and have a slight reduction in E-cadherin levels at the GSC-niche junction. Conversely, germline-specific overexpression of AdipoR inhibits natural GSC loss, suggesting that reduction in adiponectin signaling might contribute to the normal decline in GSC numbers observed over time in wild-type females. Surprisingly, AdipoR is not required for insulin sensitization of the germline, leading us to speculate that insulin sensitization is a more recently acquired function than stem cell regulation in the evolutionary history of adiponectin signaling. Our findings establish Drosophila female GSCs as a new system for future studies addressing the molecular mechanisms whereby adiponectin receptor signaling modulates stem cell fate.
Collapse
Affiliation(s)
- Kaitlin M Laws
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Baltimore, MD, USA
| | - Leesa L Sampson
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Baltimore, MD, USA
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Baltimore, MD, USA; Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
36
|
Abstract
The concept of skeletal muscle myopathy as a main determinant of exercise intolerance in chronic heart failure (HF) is gaining acceptance. Symptoms that typify HF patients, including shortness of breath and fatigue, are often directly related to the abnormalities of the skeletal muscle in HF. Besides muscular wasting, alterations in skeletal muscle energy metabolism, including insulin resistance, have been implicated in HF. Adiponectin, an adipocytokine with insulin-sensitizing properties, receives increasing interest in HF. Circulating adiponectin levels are elevated in HF patients, but high levels are paradoxically associated with poor outcome. Previous analysis of m. vastus lateralis biopsies in HF patients highlighted a striking functional adiponectin resistance. Together with increased circulating adiponectin levels, adiponectin expression within the skeletal muscle is elevated in HF patients, whereas the expression of the main adiponectin receptor and genes involved in the downstream pathway of lipid and glucose metabolism is downregulated. In addition, the adiponectin-related metabolic disturbances strongly correlate with aerobic capacity (VO2 peak), sub-maximal exercise performance and muscle strength. These observations strengthen our hypothesis that adiponectin and its receptors play a key role in the development and progression of the "heart failure myopathy". The question whether adiponectin exerts beneficial rather than detrimental effects in HF is still left unanswered. This current research overview will elucidate the emerging role of adiponectin in HF and suggests potential therapeutic targets to tackle energy wasting in these patients.
Collapse
|
37
|
Herault F, Vincent A, Dameron O, Le Roy P, Cherel P, Damon M. The Longissimus and Semimembranosus muscles display marked differences in their gene expression profiles in pig. PLoS One 2014; 9:e96491. [PMID: 24809746 PMCID: PMC4014511 DOI: 10.1371/journal.pone.0096491] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/09/2014] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Meat quality depends on skeletal muscle structure and metabolic properties. While most studies carried on pigs focus on the Longissimus muscle (LM) for fresh meat consumption, Semimembranosus (SM) is also of interest because of its importance for cooked ham production. Even if both muscles are classified as glycolytic muscles, they exhibit dissimilar myofiber composition and metabolic characteristics. The comparison of LM and SM transcriptome profiles undertaken in this study may thus clarify the biological events underlying their phenotypic differences which might influence several meat quality traits. METHODOLOGY/PRINCIPAL FINDINGS Muscular transcriptome analyses were performed using a custom pig muscle microarray: the 15 K Genmascqchip. A total of 3823 genes were differentially expressed between the two muscles (Benjamini-Hochberg adjusted P value ≤0.05), out of which 1690 and 2133 were overrepresented in LM and SM respectively. The microarray data were validated using the expression level of seven differentially expressed genes quantified by real-time RT-PCR. A set of 1047 differentially expressed genes with a muscle fold change ratio above 1.5 was used for functional characterization. Functional annotation emphasized five main clusters associated to transcriptome muscle differences. These five clusters were related to energy metabolism, cell cycle, gene expression, anatomical structure development and signal transduction/immune response. CONCLUSIONS/SIGNIFICANCE This study revealed strong transcriptome differences between LM and SM. These results suggest that skeletal muscle discrepancies might arise essentially from different post-natal myogenic activities.
Collapse
Affiliation(s)
- Frederic Herault
- INRA, UMR1348, PEGASE, F-35590 Saint-Gilles, France
- Agrocampus Ouest, UMR1348, PEGASE, F-35000 Rennes, France
| | - Annie Vincent
- INRA, UMR1348, PEGASE, F-35590 Saint-Gilles, France
- Agrocampus Ouest, UMR1348, PEGASE, F-35000 Rennes, France
| | - Olivier Dameron
- Université de Rennes1, F-35000 Rennes, France
- IRISA team Dyliss, F-35000 Rennes, France
| | - Pascale Le Roy
- INRA, UMR1348, PEGASE, F-35590 Saint-Gilles, France
- Agrocampus Ouest, UMR1348, PEGASE, F-35000 Rennes, France
| | - Pierre Cherel
- iBV-institut de Biologie Valrose, Université Nice-Sophia Antipolis UMR CNRS 7277 Inserm U1091, Parc Valrose, F-06108 Nice, France
| | - Marie Damon
- INRA, UMR1348, PEGASE, F-35590 Saint-Gilles, France
- Agrocampus Ouest, UMR1348, PEGASE, F-35000 Rennes, France
| |
Collapse
|
38
|
Fiaschi T, Magherini F, Gamberi T, Modesti PA, Modesti A. Adiponectin as a tissue regenerating hormone: more than a metabolic function. Cell Mol Life Sci 2014; 71:1917-25. [PMID: 24322911 PMCID: PMC11113778 DOI: 10.1007/s00018-013-1537-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 11/24/2013] [Accepted: 11/25/2013] [Indexed: 02/04/2023]
Abstract
The great interest that scientists have for adiponectin is primarily due to its central metabolic role. Indeed, the major function of this adipokine is the control of glucose homeostasis that it exerts regulating liver and muscle metabolism. Adiponectin has insulin-sensitizing action and leads to down-regulation of hepatic gluconeogenesis and an increase of fatty acid oxidation. In addition, adiponectin is reported to play an important role in the inhibition of inflammation. The hormone is secreted in full-length form, which can either assemble into complexes or be converted into globular form by proteolytic cleavage. Over the past few years, emerging publications reveal a more varied and pleiotropic action of this hormone. Many studies emphasize a key role of adiponectin during tissue regeneration and show that adiponectin deficiency greatly inhibits the mechanisms underlying tissue renewal. This review deals with the role of adiponectin in tissue regeneration, mainly referring to skeletal muscle regeneration, a process in which adiponectin is deeply involved. In this tissue, globular adiponectin increases proliferation, migration and myogenic properties of both resident stem cells (namely satellite cells) and non-resident muscle precursors (namely mesoangioblasts). Furthermore, skeletal muscle could be a site for the local production of the globular form that occurs in an inflamed environment. Overall, these recent findings contribute to highlight an intriguing function of adiponectin in addition to its well-recognized metabolic action.
Collapse
Affiliation(s)
- Tania Fiaschi
- Dipartimento di Scienze Biomediche, Sperimentali e Cliniche, Universita' degli Studi di Firenze, Viale Morgagni 50, 50134, Florence, Italy,
| | | | | | | | | |
Collapse
|
39
|
Vetvik KK, Sonerud T, Lindeberg M, Lüders T, Størkson RH, Jonsdottir K, Frengen E, Pietiläinen KH, Bukholm I. Globular adiponectin and its downstream target genes are up-regulated locally in human colorectal tumors: ex vivo and in vitro studies. Metabolism 2014; 63:672-81. [PMID: 24636346 DOI: 10.1016/j.metabol.2014.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 01/26/2014] [Accepted: 02/03/2014] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Low plasma adiponectin levels are linked to obesity, insulin resistance, and the risk of several types of malignancy. Despite the decline in circulating adiponectin concentrations, the increase in the expression of adiponectin receptors AdipoR1 and AdipoR2 is greater in cancerous than in normal colonic tissue. The purpose of this study was to obtain new information regarding local adiponectin signaling in the pathogenesis of colorectal cancer (CRC). METHODS We characterized the expressions of adiponectin and several of its downstream targets in paired samples of tumor tissue and adjacent noncancerous mucosa in 60 surgical patients with colorectal adenocarcinomas. RESULTS Adiponectin was expressed in both colorectal tumors and the adjacent mucosa. The expressions of adiponectin mRNA and its globular protein variant (gAd), adiponectin receptor type 1 and 5' AMP-activated protein kinase (AMPK) mRNA were significantly higher in colorectal tumors than in the adjacent mucosa. This finding was accompanied by increased mRNA expression of genes encoding proteins involved in fatty-acid trafficking and oxidation. The potential interference between adiponectin stimulation and AMPK activation through AMPK1 was examined in an in vitro model with the aid of silencing-RNA experiments. Furthermore, AMPK mRNA expression on tumors was positively correlated with a more advanced tumor stage in the patients. CONCLUSION We propose that the globular adiponectin-AMPK pathway functions in an autocrine manner in colorectal tumors, explaining some of the beneficial changes in cellular oxidative capacity in tumors in favor of tumorigenesis.
Collapse
Affiliation(s)
- Katja Kannisto Vetvik
- Department of Clinical Molecular Biology and Laboratory Sciences (EpiGen), Institute for Clinical Medicine, University of Oslo, Norway; Surgical Department, Akershus University Hospital, Lørenskog, Norway.
| | - Tonje Sonerud
- Department of Clinical Molecular Biology and Laboratory Sciences (EpiGen), Institute for Clinical Medicine, University of Oslo, Norway
| | - Mona Lindeberg
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, University of Oslo, Norway
| | - Torben Lüders
- Department of Clinical Molecular Biology and Laboratory Sciences (EpiGen), Institute for Clinical Medicine, University of Oslo, Norway
| | - Ragnhild H Størkson
- Department of Clinical Molecular Biology and Laboratory Sciences (EpiGen), Institute for Clinical Medicine, University of Oslo, Norway; Surgical Department, Østfold Hospital Trust, Fredrikstad, Norway
| | - Kristin Jonsdottir
- Department of Clinical Molecular Biology and Laboratory Sciences (EpiGen), Institute for Clinical Medicine, University of Oslo, Norway
| | - Eirik Frengen
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; Department of Medical Genetics, University of Oslo, Norway
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Department of Medicine, Division of Endocrinology, Helsinki University Central Hospital and University of Helsinki, Finland; Institute of Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Ida Bukholm
- Department of Clinical Molecular Biology and Laboratory Sciences (EpiGen), Institute for Clinical Medicine, University of Oslo, Norway; Surgical Department, Akershus University Hospital, Lørenskog, Norway; Institute of Health Promotion, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
40
|
Abstract
The beneficial metabolic effects of adiponectin which confer insulin-sensitizing and anti-diabetic effects are well established. Skeletal muscle is an important target tissue for adiponectin where it regulates glucose and fatty acid metabolism directly and via insulin sensitizing effects. Cell surface receptors and the intracellular signaling events via which adiponectin orchestrates metabolism are now becoming well characterized. The initially accepted dogma of adiponectin action was that the physiological effects were mediated via endocrine effects of adipose-derived adiponectin. However, in recent years it has been established that skeletal muscle can also produce and secrete adiponectin that can elicit important functional effects. There is evidence that skeletal muscle adiponectin resistance may develop in obesity and play a role in the pathogenesis of diabetes. In summary, adiponectin acting in an autocrine and endocrine manner has important metabolic and insulin sensitizing effects on skeletal muscle which contribute to the overall anti-diabetic outcome of adiponectin action.
Collapse
Affiliation(s)
- Ying Liu
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
41
|
Goto A, Ohno Y, Ikuta A, Suzuki M, Ohira T, Egawa T, Sugiura T, Yoshioka T, Ohira Y, Goto K. Up-regulation of adiponectin expression in antigravitational soleus muscle in response to unloading followed by reloading, and functional overloading in mice. PLoS One 2013; 8:e81929. [PMID: 24324732 PMCID: PMC3855747 DOI: 10.1371/journal.pone.0081929] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 10/28/2013] [Indexed: 12/28/2022] Open
Abstract
The purpose of this study was to investigate the expression level of adiponectin and its related molecules in hypertrophied and atrophied skeletal muscle in mice. The expression was also evaluated in C2C12 myoblasts and myotubes. Both mRNA and protein expression of adiponectin, mRNA expression of adiponectin receptor (AdipoR) 1 and AdipoR2, and protein expression of adaptor protein containing pleckstrin homology domain, phosphotyrosine binding domain, and leucine zipper motif 1 (APPL1) were observed in C2C12 myoblasts. The expression levels of these molecules in myotubes were higher than those in myoblasts. The expression of adiponectin-related molecules in soleus muscle was observed at mRNA (adiponectin, AdipoR1, AdipoR2) and protein (adiponectin, APPL1) levels. The protein expression levels of adiponectin and APPL1 were up-regulated by 3 weeks of functional overloading. Down-regulation of AdipoR1 mRNA, but not AdipoR2 mRNA, was observed in atrophied soleus muscle. The expression of adiponectin protein, AdipoR1 mRNA, and APPL1 protein was up-regulated during regrowth of unloading-associated atrophied soleus muscle. Mechanical loading, which could increase skeletal muscle mass, might be a useful stimulus for the up-regulations of adiponectin and its related molecules in skeletal muscle.
Collapse
Affiliation(s)
- Ayumi Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
| | - Yoshitaka Ohno
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
| | - Akihiro Ikuta
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
| | - Miho Suzuki
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
| | - Tomotaka Ohira
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
| | - Tatsuro Egawa
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
- Research Fellow of the Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takao Sugiura
- Department of Exercise and Health Sciences, Yamaguchi University, Yamaguchi, Japan
| | | | | | - Katsumasa Goto
- Department of Physiology, Graduate School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
- Laboratory of Physiology, School of Health Sciences, Toyohashi SOZO University, Aichi, Japan
- * E-mail:
| |
Collapse
|
42
|
Will K, Kuzinski J, Kalbe C, Palin MF, Rehfeldt C. Effects of leptin and adiponectin on the growth of porcine myoblasts are associated with changes in p44/42 MAPK signaling. Domest Anim Endocrinol 2013; 45:196-205. [PMID: 24209504 DOI: 10.1016/j.domaniend.2013.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/15/2022]
Abstract
We hypothesized that both adiponectin and leptin affect the growth of porcine skeletal muscle cells, with fatty acids acting as modifiers in adipokine action and that both adipokines influence the gene expression of their receptors. Therefore, the objective of this study was to investigate the effects of recombinant adiponectin and leptin on cell number (DNA) and DNA synthesis rate with and without oleic acid supplementation, on cell death, and on key intracellular signaling molecules of proliferating porcine myoblasts in vitro. Moreover, the mRNA expression of genes encoding for the leptin and adiponectin receptors (LEPR, ADIPOR1, ADIPOR2) as affected by leptin or adiponectin was examined. Recombinant porcine adiponectin (40 μg/mL) and leptin (20 ng/mL) increased DNA synthesis rate, measured as [(3)H]-thymidine incorporation (P < 0.01), reduced cell viability in terms of lactate dehydrogenase release (P < 0.05), or lowered DNA content after 24 h (P < 0.05). In adiponectin-treated cultures, oleic acid supplementation increased DNA synthesis rate and reduced cell number in a dose-dependent manner (P < 0.05). Both adiponectin (P = 0.07) and leptin (P < 0.05) induced a transient activation of p44/42 mitogen-activated protein kinase (MAPK) after 15 min, followed by decreases after 60 and 180 min (P < 0.05). Adiponectin tended to increase c-fos activation (P = 0.08) and decreased p53 activation at 180 min (P = 0.03). Both adiponectin and leptin down-regulated the abundance of ADIPOR2 mRNA and, transiently, of LEPR mRNA (P < 0.05). In conclusion, adiponectin and leptin may adversely affect the growth of porcine myoblasts, which is related to p44/42 MAPK signaling and associated with changes in ligand receptor gene expression.
Collapse
Affiliation(s)
- K Will
- Institute for Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | | | | | | | | |
Collapse
|
43
|
Takeda Y, Nakanishi K, Tachibana I, Kumanogoh A. Adiponectin: a novel link between adipocytes and COPD. VITAMINS AND HORMONES 2013; 90:419-35. [PMID: 23017725 DOI: 10.1016/b978-0-12-398313-8.00016-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Adiponectin (APN) is a unique adipokine with multiple salutary effects such as antiapoptotic, anti-inflammatory, and anti-oxidative activities in numerous organs and cells. Chronic obstructive pulmonary disease (COPD), a growing cause of mortality and morbidity worldwide, often results from the smoking habit and is considered a lifestyle-related disease. COPD is frequently complicated with comorbidities, such as cardiovascular disease, diabetes mellitus, and osteoporosis; however, the molecular mechanisms linking COPD and the associated comorbidities are poorly understood. Recent data have revealed a role for APN in the lung; mice lacking APN spontaneously develop a COPD-like phenotype with extrapulmonary effects, including systemic inflammation, body weight loss, and osteoporosis. This finding highlights the key role of APN in lung pathology and the novel cross talk between lung and adipose tissues. This review summarizes recent advances in understanding the physiological and pathological role of APN in the lung.
Collapse
Affiliation(s)
- Yoshito Takeda
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | | | | | | |
Collapse
|
44
|
Sánchez-Gurmaches J, Cruz-Garcia L, Gutiérrez J, Navarro I. Adiponectin effects and gene expression in rainbow trout: an in vivo and in vitro approach. ACTA ACUST UNITED AC 2012; 215:1373-83. [PMID: 22442376 DOI: 10.1242/jeb.061697] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Here we present the presence of adiponectin and adiponectin receptors [type 1 (adipoR1) and type 2 (adipoR2)] in rainbow trout (Oncorhynchus mykiss) tissues and cell cultures together with the response to different scenarios. In response to fasting, adiponectin expression was up-regulated in adipose tissue, while the expression of its receptors increased in white and red muscle. Insulin injection decreased adipoR1 expression in white and red muscles. We deduce that the adipoRs in trout muscle show opposite responses to increasing insulin plasma levels, which may maintain sensitivity to insulin in this tissue. Adiponectin expression was inhibited by the inflammatory effect of lipopolysaccharide (LPS) in adipose tissue and red muscle. Moreover, results indicate that LPS may lead to mobilization of fat reserves, increasing adipoR1 expression in adipose tissue. The effects of LPS could be mediated through tumour necrosis factor α (TNFα), at least in red muscle. Insulin, growth hormone and TNFα all diminished expression of adipoR2 in adipocytes and adipoR1 in myotubes, while insulin increased the expression of adipoR2 in the muscle cells. Adiponectin activates Akt in rainbow trout myotubes, which may lead to an increase in fatty acid uptake and oxidation. Overall, our results show that the adiponectin system responds differently to various physiological challenges and that it is hormonally controlled in vivo and in vitro. To the best of our knowledge, this is the first time this has been demonstrated in teleosts, and it may be a valuable contribution to our understanding of adipokines in fish.
Collapse
Affiliation(s)
- Juan Sánchez-Gurmaches
- Departament de Fisiologia, Facultat de Biologia, Universitat de Barcelona, Av. Diagonal 643, E-08028 Barcelona, Catalunya, Spain.
| | | | | | | |
Collapse
|
45
|
Adiponectin and skeletal muscle: pathophysiological implications in metabolic stress. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:245-56. [PMID: 22658482 DOI: 10.1016/j.ajpath.2012.03.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 03/26/2012] [Accepted: 03/29/2012] [Indexed: 12/17/2022]
Abstract
Upregulation of muscular adiponectin could act as a local protective mechanism to counteract cellular damage in obesity by weakening inflammation, oxidative stress, and apoptosis. To test this hypothesis, adiponectin-knockout (KO) and wild-type (WT) mice were fed a Western diet (WD). WT mice under WD conditions displayed 63% higher adiponectin expression in myocytes than those under standard laboratory diet (SLD) conditions (P = 0.011). WD-fed KO mice exhibited approximately threefold larger myocyte degeneration than WT mice (P = 0.003). Even under SLD conditions, myotubes of KO mice displayed already moderate immunolabeling for markers of oxidative stress (peroxiredoxin-3/5) and for a lipid peroxidation product (hydroxynonenal). Expression of tumor necrosis factor-α (TNF-α) and caspase-6, a marker of apoptosis, was also present. After WD challenge, immunoreactivity for these markers was strong in muscle of KO mice, although it was detected to a lesser extent in WT mice. Activation of NF-κB and caspase-6 doubled in myocytes of WD-fed KO mice when compared to WT mice (P < 0.001). Furthermore, muscle electrotransfer of the adiponectin gene prevented these abnormalities in WD-fed KO mice. Finally, gene abrogation of the adiponectin receptor 1 (AdipoR1) by siRNA recapitulated a pro-inflammatory state in C2C12 myotubes. Thus, upregulation of muscular adiponectin may be triggered by obesity and be crucial locally to counteract oxidative stress, inflammation, and apoptosis. These effects operate in an autocrine/paracrine manner via AdipoR1 and down-regulation of NF-κB signaling.
Collapse
|
46
|
Fiaschi T, Giannoni E, Taddei ML, Chiarugi P. Globular adiponectin activates motility and regenerative traits of muscle satellite cells. PLoS One 2012; 7:e34782. [PMID: 22629295 PMCID: PMC3356356 DOI: 10.1371/journal.pone.0034782] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 03/09/2012] [Indexed: 12/31/2022] Open
Abstract
Regeneration of adult injured skeletal muscle is due to activation of satellite cells, a population of stem cells resident beneath the basal lamina. Thus, information on soluble factors affecting satellite cell activation, as well as migration towards injury and fusion into new myofibers are essential. Here, we show that globular adiponectin (gAd), positively affects several features of muscle satellite cells. gAd activates satellite cells to exit quiescence and increases their recruitment towards myotubes. gAd elicits in satellite cells a specific motility program, involving activation of the small GTPase Rac1, as well as expression of Snail and Twist transcription factors driving a proteolytic motility, useful to reach the site of injury. We show that satellite cells produce autocrine full length adiponectin (fAd), which is converted to gAd by activated macrophages. In turns, gAd concurs to attract to the site of injury both satellite cells and macrophages and induces myogenesis in muscle satellite cells. Thus, these findings add a further role for gAd in skeletal muscle, including the hormone among factors participating in muscle regeneration.
Collapse
Affiliation(s)
- Tania Fiaschi
- Department of Biochemical Science, University of Florence, Florence, Italy
| | - Elisa Giannoni
- Department of Biochemical Science, University of Florence, Florence, Italy
| | | | - Paola Chiarugi
- Department of Biochemical Science, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
47
|
|
48
|
Dadson K, Liu Y, Sweeney G. Adiponectin action: a combination of endocrine and autocrine/paracrine effects. Front Endocrinol (Lausanne) 2011; 2:62. [PMID: 22649379 PMCID: PMC3355882 DOI: 10.3389/fendo.2011.00062] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 10/10/2011] [Indexed: 12/15/2022] Open
Abstract
The widespread physiological actions of adiponectin have now been well characterized as clinical studies and works in animal models have established strong correlations between circulating adiponectin level and various disease-related outcomes. Thus, conventional thinking attributes many of adiponectin's beneficial effects to endocrine actions of adipose-derived adiponectin. However, it is now clear that several tissues can themselves produce adiponectin and there is growing evidence that locally produced adiponectin can mediate functionally important autocrine or paracrine effects. In this review article we discuss regulation of adiponectin production, its mechanism of action via receptor isoforms and signaling pathways, and its principal physiological effects (i.e., metabolic and cardiovascular). The role of endocrine actions of adiponectin and changes in local production of adiponectin or its receptors in whole body physiology is discussed.
Collapse
Affiliation(s)
- Keith Dadson
- Department of Biology, York UniversityToronto, ON, Canada
| | - Ying Liu
- Department of Biology, York UniversityToronto, ON, Canada
| | - Gary Sweeney
- Department of Biology, York UniversityToronto, ON, Canada
- Institut Pasteur KoreaSeoul, South Korea
| |
Collapse
|
49
|
Van Berendoncks AM, Conraads VM. Functional adiponectin resistance and exercise intolerance in heart failure. Curr Heart Fail Rep 2011; 8:113-22. [PMID: 21424675 DOI: 10.1007/s11897-011-0056-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The contribution of skeletal muscle myopathy to the phenotype of patients with chronic heart failure (CHF) has become generally accepted. Besides the macro- and microscopic changes that develop during the progressive process of muscular wasting, functional abnormalities manifest in an earlier stage. Analogous to the failing heart, alterations in skeletal muscle energy metabolism, including insulin resistance, are increasingly recognized. In the search for factors causing this observed myopathy, adipokines receive growing attention. In particular, adiponectin is of special interest due to its fundamental role in skeletal muscle energy metabolism. In strong contrast with patients at risk for cardiovascular disease, circulating adiponectin levels are increased in patients with CHF, and this finding is associated with adverse outcome. Recently, the concept of functional skeletal muscle adiponectin resistance has been suggested to explain compensatory elevated adiponectin levels in CHF. Unraveling of adiponectin's complex downstream signalling pathways and insights into the concept of adiponectin resistance hopefully will disengage the road for targeted therapeutic interventions.
Collapse
Affiliation(s)
- An M Van Berendoncks
- Department of Cardiology and Laboratory for Cellular and Molecular Cardiology, Antwerp University Hospital, Wilrijkstraat 10, Edegem 2650, Belgium.
| | | |
Collapse
|
50
|
Benaitreau D, Santos ED, Leneveu MC, De Mazancourt P, Pecquery R, Dieudonné MN. Adiponectin promotes syncytialisation of BeWo cell line and primary trophoblast cells. Reprod Biol Endocrinol 2010; 8:128. [PMID: 21034435 PMCID: PMC2987948 DOI: 10.1186/1477-7827-8-128] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 10/29/2010] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND In human pregnancy, a correct placentation depends on trophoblast proliferation, differentiation, migration and invasion. These processes are highly regulated by placental hormones, growth factors and cytokines. Recently, we have shown that adiponectin, an adipokine, has anti-proliferative effects on trophoblastic cells. Here, we complete this study by demonstrating that adiponectin modulates BeWo and human villous cytotrophoblast cell differentiation. RESULTS We showed that hCG secretion was up-regulated by adiponectin treatment in both BeWo cells and human cytotrophoblasts from very early placentas (5-6 weeks). The expression of two trophoblast differentiation markers, leptin and syncytin 2, was also up-regulated by adiponectin in BeWo cells. Moreover, adiponectin treatment induced a loss of E-cadherin staining in these cells. In parallel, we demonstrated that AdipoR1 and AdipoR2 are up-regulated during forskolin induced BeWo cell differentiation, reinforcing the role of adiponectin in trophoblast syncytialization. SiRNA mediated down-regulation of AdipoR1 and AdipoR2 was used to demonstrate that adiponectin effects on differentiation were essentially mediated by these receptors. Finally, using a specific inhibitor, we demonstrated that the PKA signalling pathway could be one pathway involved in adiponectin effects on trophoblast differentiation. CONCLUSION Adiponectin enhances the differentiation process of trophoblast cells and could thus be involved in functional syncytiotrophoblast formation.
Collapse
Affiliation(s)
- Delphine Benaitreau
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire, UPRES-EA 2493, Faculté de Médecine Paris-Ile de France Ouest, PRES Universud Paris, Centre Hospitalier de Poissy-Saint Germain, 78303 Poissy Cedex, France
| | - Esther Dos Santos
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire, UPRES-EA 2493, Faculté de Médecine Paris-Ile de France Ouest, PRES Universud Paris, Centre Hospitalier de Poissy-Saint Germain, 78303 Poissy Cedex, France
| | - Marie-Christine Leneveu
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire, UPRES-EA 2493, Faculté de Médecine Paris-Ile de France Ouest, PRES Universud Paris, Centre Hospitalier de Poissy-Saint Germain, 78303 Poissy Cedex, France
| | - Philippe De Mazancourt
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire, UPRES-EA 2493, Faculté de Médecine Paris-Ile de France Ouest, PRES Universud Paris, Centre Hospitalier de Poissy-Saint Germain, 78303 Poissy Cedex, France
| | - René Pecquery
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire, UPRES-EA 2493, Faculté de Médecine Paris-Ile de France Ouest, PRES Universud Paris, Centre Hospitalier de Poissy-Saint Germain, 78303 Poissy Cedex, France
| | - Marie-Noëlle Dieudonné
- Université de Versailles-St Quentin, Service de Biochimie et Biologie Moléculaire, UPRES-EA 2493, Faculté de Médecine Paris-Ile de France Ouest, PRES Universud Paris, Centre Hospitalier de Poissy-Saint Germain, 78303 Poissy Cedex, France
| |
Collapse
|