1
|
Gao Y, Chen Q, Wu Z, Yuan L. Regulation of pancreatic β cells by exosomes from different sources. Diabetes Res Clin Pract 2025:112222. [PMID: 40324722 DOI: 10.1016/j.diabres.2025.112222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Diabetes is a chronic metabolic disorder with rising global prevalence, particularly in developed and high-income regions. Central to its pathogenesis is the dysfunction of pancreatic β-cells, alongside impaired glucose and lipid metabolism in peripheral insulin-responsive tissues. Exosomes are nano-sized extracellular vesicles essential for intercellular communication and have emerged as pivotal regulators of metabolic homeostasis. Secreted by virtually all cell types, exosomes encapsulate bioactive cargo that reflects their cellular origin and physiological state, thereby exerting diverse functional effects. Recent evidence highlights the role of exosomes derived from the liver, gut, adipose tissue, skeletal muscle, and mesenchymal stem cells in modulating β-cell proliferation, insulin secretion, and survival. In peripheral tissues exosomes also influence insulin sensitivity by regulating glucose and lipid metabolism, ultimately shaping β-cell responses under hyperglycemic conditions. A more comprehensive understanding of exosome-mediated crosstalk between metabolic organs and pancreatic β-cells could pave the way for the development of exosome-based diagnostic tools and therapeutic strategies aimed at improving early detection, prevention, and treatment of the diabetes.
Collapse
Affiliation(s)
- Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhuoying Wu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
2
|
Orioli L, Thissen JP. Myokines as potential mediators of changes in glucose homeostasis and muscle mass after bariatric surgery. Front Endocrinol (Lausanne) 2025; 16:1554617. [PMID: 40171198 PMCID: PMC11958187 DOI: 10.3389/fendo.2025.1554617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Myokines are bioactive peptides released by skeletal muscle. Myokines exert auto-, para-, or endocrine effects, enabling them to regulate many aspects of metabolism in various tissues. However, the contribution of myokines to the dramatic changes in glucose homeostasis and muscle mass induced by bariatric surgery has not been established. Our review highlights that myokines such as brain-derived neurotrophic factor (BDNF), meteorin-like protein (Metrnl), secreted protein acidic and rich in cysteine (SPARC), apelin (APLN) and myostatin (MSTN) may mediate changes in glucose homeostasis and muscle mass after bariatric surgery. Our review also identifies myonectin as an interesting candidate for future studies, as this myokine may regulate lipid metabolism and muscle mass after bariatric surgery. These myokines may provide novel therapeutic targets and biomarkers for obesity, type 2 diabetes and sarcopenia.
Collapse
Affiliation(s)
- Laura Orioli
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Research Laboratory of Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
3
|
Langlois A, Cherfan J, Meugnier E, Rida A, Arous C, Peronet C, Hamdard H, Zarrouki B, Wehrle‐Haller B, Pinget M, Craige SM, Bouzakri K. DECORIN, a triceps-derived myokine, protects sorted β-cells and human islets against chronic inflammation associated with type 2 diabetes. Acta Physiol (Oxf) 2025; 241:e14267. [PMID: 39844653 PMCID: PMC11754997 DOI: 10.1111/apha.14267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/28/2024] [Accepted: 01/01/2025] [Indexed: 01/24/2025]
Abstract
AIM Pancreatic β-cells are susceptible to inflammation, leading to decreased insulin production/secretion and cell death. Previously, we have identified a novel triceps-derived myokine, DECORIN, which plays a pivotal role in skeletal muscle-to-pancreas interorgan communication. However, whether DECORIN can directly impact β-cell function and susceptibility to inflammation remains unexplored. METHODS The effect of DECORIN was assessed in sorted human and rat β-cell and human islets from healthy and type 2 diabetes (T2D) donors. We assessed glucose-stimulated insulin secretion (GSIS) and cytokine-mediated cell death. We then challenged sorted β-cells and human islets with inflammatory cytokines commonly associated with diabetes, such as tumor necrosis factor-α (TNF-α) alone or in combination with interleukin1-β (IL1-β) and interferon-γ (cytomix). RESULTS DECORIN enhanced cell spreading and the localization of phosphorylated FAK at adhesions, promoting GSIS under basal conditions. It also increased insulin granule docking adhesion length and countered the inhibitory effects of TNF-α on adhesion and actin remodeling at the β-cell surface, resulting in preserved GSIS. DECORIN protected from cell death in sorted β-cells and islets challenged with TNF-α alone or TNF-α + cytomix. Interestingly, DECORIN increased both insulin content and secretion in human islets from T2D individuals. Additionally, DECORIN treatment reversed the impaired gene expression caused by T2D and enhanced the expression of genes essential for islet function and metabolism. CONCLUSION Collectively, we have shown that DECORIN had a beneficial effect on human islets, protecting them from inflammation-induced cell death. In T2D islets, DECORIN restores islet function and reverses the expression of T2D-associated genes. Based on our data, we propose that DECORIN is a promising therapeutic target for diabetes-associated inflammation and diabetes itself.
Collapse
Affiliation(s)
- Allan Langlois
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Julien Cherfan
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Emmanuelle Meugnier
- CarMeN Laboratory, Inserm U1060, INRAE UMR1397, Univ‐LyonUniversité Claude Bernard Lyon‐1LyonFrance
| | - Ahmad Rida
- ILONOV, Boulevard René LericheStrasbourgFrance
| | - Caroline Arous
- Department of Cell Physiology and Metabolism, Centre Médical UniversitaireUniversity of GenevaGenevaSwitzerland
| | - Claude Peronet
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Harzo Hamdard
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
| | - Bader Zarrouki
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolic (CVRM), BioPharmaceuticals R&DGothenburgSweden
| | - Bernhard Wehrle‐Haller
- Department of Cell Physiology and Metabolism, Centre Médical UniversitaireUniversity of GenevaGenevaSwitzerland
| | - Michel Pinget
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
- ILONOV, Boulevard René LericheStrasbourgFrance
| | - Siobhan M. Craige
- Department of Human Nutrition, Foods, and ExerciseVirginia TechBlacksburgVirginiaUSA
| | - Karim Bouzakri
- UR Diabète et Thérapeutiques, Centre européen d'étude du DiabèteUniversité de StrasbourgStrasbourgFrance
- ILONOV, Boulevard René LericheStrasbourgFrance
| |
Collapse
|
4
|
Calcaterra V, Magenes VC, Bianchi A, Rossi V, Gatti A, Marin L, Vandoni M, Zuccotti G. How Can Promoting Skeletal Muscle Health and Exercise in Children and Adolescents Prevent Insulin Resistance and Type 2 Diabetes? Life (Basel) 2024; 14:1198. [PMID: 39337980 PMCID: PMC11433096 DOI: 10.3390/life14091198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Skeletal muscle secretome, through its paracrine and endocrine functions, contributes to the maintenance and regulation of overall physiological health. We conducted a narrative review on the role of skeletal muscle and exercise in maintaining glucose homeostasis, driving insulin resistance (IR), and preventing type 2 diabetes in pediatric populations, especially in the context of overweight and obesity. Myokines such as interleukin (IL)-6, IL-8, and IL-15, as well as irisin, myonectin, and myostatin, appear to play a crucial role in IR. Skeletal muscle can also become a target of obesity-induced and IR-induced inflammation. In the correlation between muscle, IR, and inflammation, the role of infiltration of the immune cells and the microvasculature may also be considered. It remains unclear which exercise approach is the best; however, combining aerobic exercise with resistance training seems to be the most effective strategy for managing IR, with high-intensity activities offering superior metabolic benefits and long-term adherence. Encouraging daily participation in enjoyable and engaging exercise is key for long-term commitment and effective glucose metabolism management. Promoting physical activity in children and adolescents must be a top priority for public health, not only in terms of individual quality of life and well-being but also for community health.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100 Pavia, Italy
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Vittoria Carlotta Magenes
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Alice Bianchi
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Virginia Rossi
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
| | - Alessandro Gatti
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy; (A.G.); (L.M.); (M.V.)
| | - Luca Marin
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy; (A.G.); (L.M.); (M.V.)
| | - Matteo Vandoni
- Laboratory of Adapted Motor Activity (LAMA), Department of Public Health, Experimental Medicine and Forensic Science, University of Pavia, 27100 Pavia, Italy; (A.G.); (L.M.); (M.V.)
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (V.C.M.); (A.B.); (V.R.); (G.Z.)
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milano, Italy
| |
Collapse
|
5
|
Marcos-Pérez D, Cruces-Salguero S, García-Domínguez E, Araúzo-Bravo MJ, Gómez-Cabrera MC, Viña J, Vergara I, Matheu A. Physical Interventions Restore Physical Frailty and the Expression of CXCL-10 and IL-1β Inflammatory Biomarkers in Old Individuals and Mice. Biomolecules 2024; 14:166. [PMID: 38397403 PMCID: PMC10886745 DOI: 10.3390/biom14020166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/15/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Frailty is a geriatric syndrome associated with negative health outcomes that represents a dynamic condition with a potential of reversibility after physical exercise interventions. Typically, inflammatory and senescence markers are increased in frail individuals. However, the impact that physical exercise exerts on inflammatory and senescence biomarkers remains unknown. We assessed the effect of physical intervention in old individuals and mice and determined the expression of inflammatory and senescence markers. METHODS Twelve elderly individuals were enrolled from a primary care setting to a 3-month intervention. Frailty was measured by SPPB and the expression of biomarkers by cytokine array and RT-qPCR. In addition, 12 aged C57BL/6 mice completed an intervention, and inflammation and senescence markers were studied. RESULTS The physical intervention improved the SPPB score, reducing frail and pre-frail individuals. This was correlated with a reduction in several pro-inflammatory biomarkers such as IL-6, CXCL-1, CXCL-10, IL-1β, IL-7, GM-CSF as well as p16INK4a and p21CIP1 senescence markers. Otherwise, the levels of anti-inflammatory biomarker IL-4 were significantly increased. Moreover, the physical intervention in mice also improved their functional capacity and restored the expression of inflammatory (Il-1β, Cxcl-10, Il-6, and Cxcl-1) and senescence (p21Cip1) markers. Additionally, PLSDA and ROC curve analysis revealed CXCL-10 and IL-1β to be the biomarkers of functional improvement in both cohorts. CONCLUSIONS Our results showed that a physical intervention improves physical frailty, and reverses inflammation and senescence biomarkers comprising CXCL-10 and IL-1β.
Collapse
Affiliation(s)
- Diego Marcos-Pérez
- Cellular Oncology Group, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain; (D.M.-P.); (S.C.-S.)
| | - Sara Cruces-Salguero
- Cellular Oncology Group, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain; (D.M.-P.); (S.C.-S.)
| | - Esther García-Domínguez
- Freshage Research Group, Faculty of Medicine, Fundación Investigación Hospital Clínico Universitario/Health Research Institute INCLIVA, University of Valencia, 46010 Valencia, Spain; (E.G.-D.); (M.C.G.-C.); (J.V.)
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
| | - Marcos J. Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014 San Sebastián, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Mari Carmen Gómez-Cabrera
- Freshage Research Group, Faculty of Medicine, Fundación Investigación Hospital Clínico Universitario/Health Research Institute INCLIVA, University of Valencia, 46010 Valencia, Spain; (E.G.-D.); (M.C.G.-C.); (J.V.)
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
| | - José Viña
- Freshage Research Group, Faculty of Medicine, Fundación Investigación Hospital Clínico Universitario/Health Research Institute INCLIVA, University of Valencia, 46010 Valencia, Spain; (E.G.-D.); (M.C.G.-C.); (J.V.)
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
| | - Itziar Vergara
- Primary Care Group, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain;
| | - Ander Matheu
- Cellular Oncology Group, Biogipuzkoa Health Research Institute, 20014 San Sebastián, Spain; (D.M.-P.); (S.C.-S.)
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERfes), 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
6
|
Veluthakal R, Esparza D, Hoolachan JM, Balakrishnan R, Ahn M, Oh E, Jayasena CS, Thurmond DC. Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression. Int J Mol Sci 2024; 25:1504. [PMID: 38338783 PMCID: PMC10855860 DOI: 10.3390/ijms25031504] [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: 12/22/2023] [Revised: 01/15/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Type 2 diabetes (T2D) is a heterogenous disease, and conventionally, peripheral insulin resistance (IR) was thought to precede islet β-cell dysfunction, promoting progression from prediabetes to T2D. New evidence suggests that T2D-lean individuals experience early β-cell dysfunction without significant IR. Regardless of the primary event (i.e., IR vs. β-cell dysfunction) that contributes to dysglycemia, significant early-onset oxidative damage and mitochondrial dysfunction in multiple metabolic tissues may be a driver of T2D onset and progression. Oxidative stress, defined as the generation of reactive oxygen species (ROS), is mediated by hyperglycemia alone or in combination with lipids. Physiological oxidative stress promotes inter-tissue communication, while pathological oxidative stress promotes inter-tissue mis-communication, and new evidence suggests that this is mediated via extracellular vesicles (EVs), including mitochondria containing EVs. Under metabolic-related stress conditions, EV-mediated cross-talk between β-cells and skeletal muscle likely trigger mitochondrial anomalies leading to prediabetes and T2D. This article reviews the underlying molecular mechanisms in ROS-related pathogenesis of prediabetes, including mitophagy and mitochondrial dynamics due to oxidative stress. Further, this review will describe the potential of various therapeutic avenues for attenuating oxidative damage, reversing prediabetes and preventing progression to T2D.
Collapse
Affiliation(s)
- Rajakrishnan Veluthakal
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, 1500 E. Duarte Rd, Duarte, CA 91010, USA; (D.E.); (J.M.H.); (R.B.); (M.A.); (E.O.); (C.S.J.)
| | | | | | | | | | | | | | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope Beckman Research Institute, 1500 E. Duarte Rd, Duarte, CA 91010, USA; (D.E.); (J.M.H.); (R.B.); (M.A.); (E.O.); (C.S.J.)
| |
Collapse
|
7
|
Ung CY, Correia C, Li H, Adams CM, Westendorf JJ, Zhu S. Multiorgan locked-state model of chronic diseases and systems pharmacology opportunities. Drug Discov Today 2024; 29:103825. [PMID: 37967790 PMCID: PMC11109989 DOI: 10.1016/j.drudis.2023.103825] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/29/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
With increasing human life expectancy, the global medical burden of chronic diseases is growing. Hence, chronic diseases are a pressing health concern and will continue to be in decades to come. Chronic diseases often involve multiple malfunctioning organs in the body. An imminent question is how interorgan crosstalk contributes to the etiology of chronic diseases. We conceived the locked-state model (LoSM), which illustrates how interorgan communication can give rise to body-wide memory-like properties that 'lock' healthy or pathological conditions. Next, we propose cutting-edge systems biology and artificial intelligence strategies to decipher chronic multiorgan locked states. Finally, we discuss the clinical implications of the LoSM and assess the power of systems-based therapies to dismantle pathological multiorgan locked states while improving treatments for chronic diseases.
Collapse
Affiliation(s)
- Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Christopher M Adams
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jennifer J Westendorf
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA; Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Shizhen Zhu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
8
|
Xourafa G, Korbmacher M, Roden M. Inter-organ crosstalk during development and progression of type 2 diabetes mellitus. Nat Rev Endocrinol 2024; 20:27-49. [PMID: 37845351 DOI: 10.1038/s41574-023-00898-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2023] [Indexed: 10/18/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by tissue-specific insulin resistance and pancreatic β-cell dysfunction, which result from the interplay of local abnormalities within different tissues and systemic dysregulation of tissue crosstalk. The main local mechanisms comprise metabolic (lipid) signalling, altered mitochondrial metabolism with oxidative stress, endoplasmic reticulum stress and local inflammation. While the role of endocrine dysregulation in T2DM pathogenesis is well established, other forms of inter-organ crosstalk deserve closer investigation to better understand the multifactorial transition from normoglycaemia to hyperglycaemia. This narrative Review addresses the impact of certain tissue-specific messenger systems, such as metabolites, peptides and proteins and microRNAs, their secretion patterns and possible alternative transport mechanisms, such as extracellular vesicles (exosomes). The focus is on the effects of these messengers on distant organs during the development of T2DM and progression to its complications. Starting from the adipose tissue as a major organ relevant to T2DM pathophysiology, the discussion is expanded to other key tissues, such as skeletal muscle, liver, the endocrine pancreas and the intestine. Subsequently, this Review also sheds light on the potential of multimarker panels derived from these biomarkers and related multi-omics for the prediction of risk and progression of T2DM, novel diabetes mellitus subtypes and/or endotypes and T2DM-related complications.
Collapse
Affiliation(s)
- Georgia Xourafa
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Düsseldorf, Germany
| | - Melis Korbmacher
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Düsseldorf, Germany.
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
9
|
Mahmoudi-Aznaveh A, Tavoosidana G, Najmabadi H, Azizi Z, Ardestani A. The liver-derived exosomes stimulate insulin gene expression in pancreatic beta cells under condition of insulin resistance. Front Endocrinol (Lausanne) 2023; 14:1303930. [PMID: 38027137 PMCID: PMC10661932 DOI: 10.3389/fendo.2023.1303930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction An insufficient functional beta cell mass is a core pathological hallmark of type 2 diabetes (T2D). Despite the availability of several effective pharmaceuticals for diabetes management, there is an urgent need for novel medications to protect pancreatic beta cells under diabetic conditions. Integrative organ cross-communication controls the energy balance and glucose homeostasis. The liver and pancreatic islets have dynamic cross-communications where the liver can trigger a compensatory beta cell mass expansion and enhanced hormonal secretion in insulin-resistant conditions. However, the indispensable element(s) that foster beta cell proliferation and insulin secretion have yet to be completely identified. Exosomes are important extracellular vehicles (EVs) released by most cell types that transfer biological signal(s), including metabolic messengers such as miRNA and peptides, between cells and organs. Methods We investigated whether beta cells can take up liver-derived exosomes and examined their impact on beta cell functional genes and insulin expression. Exosomes isolated from human liver HepG2 cells were characterized using various methods, including Transmission Electron Microscopy (TEM), dynamic light scattering (DLS), and Western blot analysis of exosomal markers. Exosome labeling and cell uptake were assessed using CM-Dil dye. The effect of liver cell-derived exosomes on Min6 beta cells was determined through gene expression analyses of beta cell markers and insulin using qPCR, as well as Akt signaling using Western blotting. Results Treatment of Min6 beta cells with exosomes isolated from human liver HepG2 cells treated with insulin receptor antagonist S961 significantly increased the expression of beta cell markers Pdx1, NeuroD1, and Ins1 compared to the exosomes isolated from untreated cells. In line with this, the activity of AKT kinase, an integral component of the insulin receptor pathway, is elevated in pancreatic beta cells, as represented by an increase in AKT's downstream substrate, FoxO1 phosphorylation. Discussions This study suggests that liver-derived exosomes may carry a specific molecular cargo that can affect insulin expression in pancreatic beta cells, ultimately affecting glucose homeostasis.
Collapse
Affiliation(s)
- Azam Mahmoudi-Aznaveh
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Tavoosidana
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Zahra Azizi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Ardestani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| |
Collapse
|
10
|
Orioli L, Canouil M, Sawadogo K, Ning L, Deldicque L, Lause P, de Barsy M, Froguel P, Loumaye A, Deswysen Y, Navez B, Bonnefond A, Thissen JP. Identification of myokines susceptible to improve glucose homeostasis after bariatric surgery. Eur J Endocrinol 2023; 189:409-421. [PMID: 37638789 DOI: 10.1093/ejendo/lvad122] [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: 04/01/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023]
Abstract
IMPORTANCE AND OBJECTIVE The identification of myokines susceptible to improve glucose homeostasis following bariatric surgery could lead to new therapeutic approaches for type 2 diabetes. METHODS Changes in the homeostasis model assessment (HOMA) test were assessed in patients before and 3 months after bariatric surgery. Changes in myokines expression and circulating levels were assessed using real-time quantitative polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA). Myokines known to regulate glucose homeostasis were identified using literature (targeted study) and putative myokines using RNA-sequencing (untargeted study). A linear regression analysis adjusted for age and sex was used to search for associations between changes in the HOMA test and changes in myokines. RESULTS In the targeted study, brain-derived neurotrophic factor (BDNF) expression was upregulated (+30%, P = .006) while BDNF circulating levels were decreased (-12%, P = .001). Upregulated BDNF expression was associated with decreased HOMA of insulin resistance (HOMA-IR) (adjusted estimate [95% confidence interval {CI}]: -0.51 [-0.88 to -0.13], P = .010). Decreased BDNF serum levels were associated with decreased HOMA of beta-cell function (HOMA-B) (adjusted estimate [95% CI] = 0.002 [0.00002-0.0031], P = .046). In the untargeted study, upregulated putative myokines included XYLT1 (+64%, P < .001), LGR5 (+57, P< .001), and SPINK5 (+46%, P < .001). Upregulated LGR5 was associated with decreased HOMA-IR (adjusted estimate [95% CI] = -0.50 [-0.86 to -0.13], P = .009). Upregulated XYLT1 and SPINK5 were associated with increased HOMA of insulin sensitivity (HOMA-S) (respectively, adjusted estimate [95% CI] = 109.1 [28.5-189.8], P = .009 and 16.5 [0.87-32.19], P = .039). CONCLUSIONS Improved glucose homeostasis following bariatric surgery is associated with changes in myokines expression and circulating levels. In particular, upregulation of BDNF, XYLT1, SPINK5, and LGR5 is associated with improved insulin sensitivity. These results suggest that these myokines could contribute to improved glucose homeostasis following bariatric surgery. STUDY REGISTRATION NCT03341793 on ClinicalTrials.gov (https://clinicaltrials.gov/).
Collapse
Affiliation(s)
- Laura Orioli
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Mickaël Canouil
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Kiswendsida Sawadogo
- Statistical Support Unit, King Albert II Cancer and Hematology Institute, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Lijiao Ning
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Louise Deldicque
- Institute of NeuroScience, Université Catholique de Louvain, 1348 Louvain-La-Neuve, Belgium
| | - Pascale Lause
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Marie de Barsy
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Philippe Froguel
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
- Department of Metabolism, Digestion, and Reproduction, Imperial College London, London SW7 2BX, United Kingdom
| | - Audrey Loumaye
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Yannick Deswysen
- Department of Oeso-gastro-duodenal and Bariatric Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Benoit Navez
- Department of Oeso-gastro-duodenal and Bariatric Surgery, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Amélie Bonnefond
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, 59000 Lille, France
- University of Lille, Lille University Hospital, 59000 Lille, France
| | - Jean-Paul Thissen
- Endocrinology, Diabetes, and Nutrition, Institute of Experimental and Clinical Research, Université Catholique de Louvain, 1200 Brussels, Belgium
- Department of Endocrinology and Nutrition, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
11
|
Hall LG, Thyfault JP, Johnson JD. Exercise and inactivity as modifiers of β cell function and type 2 diabetes risk. J Appl Physiol (1985) 2023; 134:823-839. [PMID: 36759159 PMCID: PMC10042613 DOI: 10.1152/japplphysiol.00472.2022] [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: 08/15/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/11/2023] Open
Abstract
Exercise and regular physical activity are beneficial for the prevention and management of metabolic diseases such as obesity and type 2 diabetes, whereas exercise cessation, defined as deconditioning from regular exercise or physical activity that has lasted for a period of months to years, can lead to metabolic derangements that drive disease. Adaptations to the insulin-secreting pancreatic β-cells are an important benefit of exercise, whereas less is known about how exercise cessation affects these cells. Our aim is to review the impact that exercise and exercise cessation have on β-cell function, with a focus on the evidence from studies examining glucose-stimulated insulin secretion (GSIS) using gold-standard techniques. Potential mechanisms by which the β-cell adapts to exercise, including exerkine and incretin signaling, autonomic nervous system signaling, and changes in insulin clearance, will also be explored. We will highlight areas for future research.
Collapse
Affiliation(s)
- Liam G Hall
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - John P Thyfault
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
- KU Diabetes Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
12
|
Role of Skeletal Muscle in the Pathogenesis and Management of Type 2 Diabetes: A Special Focus on Asian Indians. J Indian Inst Sci 2023. [DOI: 10.1007/s41745-022-00349-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
13
|
Melena I, Hughes JW. Islet cilia and glucose homeostasis. Front Cell Dev Biol 2022; 10:1082193. [PMID: 36531945 PMCID: PMC9751591 DOI: 10.3389/fcell.2022.1082193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/22/2022] [Indexed: 09/05/2023] Open
Abstract
Diabetes is a growing pandemic affecting over ten percent of the U.S. population. Individuals with all types of diabetes exhibit glucose dysregulation due to altered function and coordination of pancreatic islets. Within the critical intercellular space in pancreatic islets, the primary cilium emerges as an important physical structure mediating cell-cell crosstalk and signal transduction. Many events leading to hormone secretion, including GPCR and second-messenger signaling, are spatiotemporally regulated at the level of the cilium. In this review, we summarize current knowledge of cilia action in islet hormone regulation and glucose homeostasis, focusing on newly implicated ciliary pathways that regulate insulin exocytosis and intercellular communication. We present evidence of key signaling proteins on islet cilia and discuss ways in which cilia might functionally connect islet endocrine cells with the non-endocrine compartments. These discussions aim to stimulate conversations regarding the extent of cilia-controlled glucose homeostasis in health and in metabolic diseases.
Collapse
Affiliation(s)
| | - Jing W. Hughes
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
14
|
Amir Levy Y, P Ciaraldi T, R. Mudaliar S, A. Phillips S, R. Henry R. Adipose tissue from subjects with type 2 diabetes exhibits impaired capillary formation in response to GROα: involvement of MMPs-2 and -9. Adipocyte 2022; 11:276-286. [PMID: 35481427 PMCID: PMC9116416 DOI: 10.1080/21623945.2022.2070949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 12/03/2022] Open
Abstract
Type 2 Diabetes (T2D) is associated with impaired vascularization of adipose tissue (AT) . IL8, GROα and IL15 are pro-angiogenic myokines, secreted at elevated levels by T2D myotubes. We explored the direct impact of these myokines on AT vascularization. AT explants from subjects with T2D and without diabetes (non-diabetic, ND) were treated with rIL8, rGROα and rIL15 in concentrations equal to those in conditioned media (CM) from T2D and ND myotubes, and sprout formation evaluated. Endothelial cells (EC) were isolated from T2D and ND-AT, treated with rGROα and tube formation evaluated. Finally, we investigated the involvement of MMP-2 and -9 in vascularization. ND and T2D concentrations of IL8 or IL15 caused similar stimulation of sprout formation in ND- and T2D-AT. GROα exerted a similar effect in ND-AT. When T2D-AT explants were exposed to GROα, sprout formation in response to T2D concentrations was reduced compared to ND. Exposure of EC from T2D-AT to GROα at T2D concentrations resulted in reduced tube formation. Reduced responses to GROα in T2D-AT and EC were also seen for secretion of MMP-2 and -9. The data indicate that skeletal muscle can potentially regulate AT vascularization, with T2D-AT having impairments in sensitivity to GROα, while responding normally to IL8 and IL15.
Collapse
Affiliation(s)
- Yifat Amir Levy
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| | - Theodore P Ciaraldi
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| | - Sunder R. Mudaliar
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| | - Susan A. Phillips
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Pediatrics, University of California, La Jolla, CA, USA
| | - Robert R. Henry
- Center for Metabolic Research, Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Departments of Medicine, University of California, La Jolla, CA, USA
| |
Collapse
|
15
|
Abstract
Exosomes are a class of extracellular vesicles with a diameter of 50-100 nm secreted by various cells. They are generated through complex intracellular production mechanisms before being secreted to the extracellular environment. Due to their inclusion of proteins, lipids, and nucleic acids, exosomes play an important role in intercellular communication. Pancreatic β-cells play an irreplaceable role in the body's glucose metabolism. Their dysfunction is one of the causes of diabetes. Exosomes of various cells regulate the function of β-cells by regulating autoimmunity, delivering non-coding RNAs, or directly regulating intracellular signal pathways. This communication between β-cells and other cells plays an important role in the pathogenesis and development of diabetes, and has potential for clinical application. This paper reviews the biological sources and functions of exosomes, as well as intercellular crosstalk between β-cells and other cells that is involved in β-cell failure and regeneration.
Collapse
Affiliation(s)
- Yu Wu
- Diabetes Research Center, Medical School, Ningbo University, Ningbo, China
| | - Qin Huang
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Shizhong Bu
- Diabetes Research Center, Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
16
|
Sheng CY, Son YH, Jang J, Park SJ. In vitro skeletal muscle models for type 2 diabetes. BIOPHYSICS REVIEWS 2022; 3:031306. [PMID: 36124295 PMCID: PMC9478902 DOI: 10.1063/5.0096420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Type 2 diabetes mellitus, a metabolic disorder characterized by abnormally elevated blood sugar, poses a growing social, economic, and medical burden worldwide. The skeletal muscle is the largest metabolic organ responsible for glucose homeostasis in the body, and its inability to properly uptake sugar often precedes type 2 diabetes. Although exercise is known to have preventative and therapeutic effects on type 2 diabetes, the underlying mechanism of these beneficial effects is largely unknown. Animal studies have been conducted to better understand the pathophysiology of type 2 diabetes and the positive effects of exercise on type 2 diabetes. However, the complexity of in vivo systems and the inability of animal models to fully capture human type 2 diabetes genetics and pathophysiology are two major limitations in these animal studies. Fortunately, in vitro models capable of recapitulating human genetics and physiology provide promising avenues to overcome these obstacles. This review summarizes current in vitro type 2 diabetes models with focuses on the skeletal muscle, interorgan crosstalk, and exercise. We discuss diabetes, its pathophysiology, common in vitro type 2 diabetes skeletal muscle models, interorgan crosstalk type 2 diabetes models, exercise benefits on type 2 diabetes, and in vitro type 2 diabetes models with exercise.
Collapse
Affiliation(s)
- Christina Y. Sheng
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Young Hoon Son
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | - Sung-Jin Park
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
17
|
Biondi G, Marrano N, Borrelli A, Rella M, Palma G, Calderoni I, Siciliano E, Lops P, Giorgino F, Natalicchio A. Adipose Tissue Secretion Pattern Influences β-Cell Wellness in the Transition from Obesity to Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23105522. [PMID: 35628332 PMCID: PMC9143684 DOI: 10.3390/ijms23105522] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/10/2022] Open
Abstract
The dysregulation of the β-cell functional mass, which is a reduction in the number of β-cells and their ability to secure adequate insulin secretion, represents a key mechanistic factor leading to the onset of type 2 diabetes (T2D). Obesity is recognised as a leading cause of β-cell loss and dysfunction and a risk factor for T2D. The natural history of β-cell failure in obesity-induced T2D can be divided into three steps: (1) β-cell compensatory hyperplasia and insulin hypersecretion, (2) insulin secretory dysfunction, and (3) loss of β-cell mass. Adipose tissue (AT) secretes many hormones/cytokines (adipokines) and fatty acids that can directly influence β-cell function and viability. As this secretory pattern is altered in obese and diabetic patients, it is expected that the cross-talk between AT and pancreatic β-cells could drive the maintenance of the β-cell integrity under physiological conditions and contribute to the reduction in the β-cell functional mass in a dysmetabolic state. In the current review, we summarise the evidence of the ability of the AT secretome to influence each step of β-cell failure, and attempt to draw a timeline of the alterations in the adipokine secretion pattern in the transition from obesity to T2D that reflects the progressive deterioration of the β-cell functional mass.
Collapse
|
18
|
Balakrishnan R, Thurmond DC. Mechanisms by Which Skeletal Muscle Myokines Ameliorate Insulin Resistance. Int J Mol Sci 2022; 23:4636. [PMID: 35563026 PMCID: PMC9102915 DOI: 10.3390/ijms23094636] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 12/17/2022] Open
Abstract
The skeletal muscle is the largest organ in the body and secretes circulating factors, including myokines, which are involved in various cellular signaling processes. Skeletal muscle is vital for metabolism and physiology and plays a crucial role in insulin-mediated glucose disposal. Myokines have autocrine, paracrine, and endocrine functions, serving as critical regulators of myogenic differentiation, fiber-type switching, and maintaining muscle mass. Myokines have profound effects on energy metabolism and inflammation, contributing to the pathophysiology of type 2 diabetes (T2D) and other metabolic diseases. Myokines have been shown to increase insulin sensitivity, thereby improving glucose disposal and regulating glucose and lipid metabolism. Many myokines have now been identified, and research on myokine signaling mechanisms and functions is rapidly emerging. This review summarizes the current state of the field regarding the role of myokines in tissue cross-talk, including their molecular mechanisms, and their potential as therapeutic targets for T2D.
Collapse
Affiliation(s)
| | - Debbie C. Thurmond
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope Beckman Research Institute, 1500 E. Duarte Road, Duarte, CA 91010, USA;
| |
Collapse
|
19
|
Rodríguez-Comas J, Ramón-Azcón J. Islet-on-a-chip for the study of pancreatic β-cell function. IN VITRO MODELS 2022; 1:41-57. [PMID: 39872972 PMCID: PMC11749753 DOI: 10.1007/s44164-021-00005-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/25/2021] [Accepted: 11/08/2021] [Indexed: 01/30/2025]
Abstract
Diabetes mellitus is a significant public health problem worldwide. It encompasses a group of chronic disorders characterized by hyperglycemia, resulting from pancreatic islet dysfunction or as a consequence of insulin-producing β-cell death. Organ-on-a-chip platforms have emerged as technological systems combining cell biology, engineering, and biomaterial technological advances with microfluidics to recapitulate a specific organ's physiological or pathophysiological environment. These devices offer a novel model for the screening of pharmaceutical agents and to study a particular disease. In the field of diabetes, a variety of microfluidic devices have been introduced to recreate native islet microenvironments and to understand pancreatic β-cell kinetics in vitro. This kind of platforms has been shown fundamental for the study of the islet function and to assess the quality of these islets for subsequent in vivo transplantation. However, islet physiological systems are still limited compared to other organs and tissues, evidencing the difficulty to study this "organ" and the need for further technological advances. In this review, we summarize the current state of islet-on-a-chip platforms that have been developed so far. We recapitulate the most relevant studies involving pancreatic islets and microfluidics, focusing on the molecular and cellular-scale activities that underlie pancreatic β-cell function.
Collapse
Affiliation(s)
- Júlia Rodríguez-Comas
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
| | - Javier Ramón-Azcón
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac, 10-12, 08028 Barcelona, Spain
- ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| |
Collapse
|
20
|
Fernández-Millán E, Guillén C. Multi-Organ Crosstalk with Endocrine Pancreas: A Focus on How Gut Microbiota Shapes Pancreatic Beta-Cells. Biomolecules 2022; 12:biom12010104. [PMID: 35053251 PMCID: PMC8773909 DOI: 10.3390/biom12010104] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes (T2D) results from impaired beta-cell function and insufficient beta-cell mass compensation in the setting of insulin resistance. Current therapeutic strategies focus their efforts on promoting the maintenance of functional beta-cell mass to ensure appropriate glycemic control. Thus, understanding how beta-cells communicate with metabolic and non-metabolic tissues provides a novel area for investigation and implicates the importance of inter-organ communication in the pathology of metabolic diseases such as T2D. In this review, we provide an overview of secreted factors from diverse organs and tissues that have been shown to impact beta-cell biology. Specifically, we discuss experimental and clinical evidence in support for a role of gut to beta-cell crosstalk, paying particular attention to bacteria-derived factors including short-chain fatty acids, lipopolysaccharide, and factors contained within extracellular vesicles that influence the function and/or the survival of beta cells under normal or diabetogenic conditions.
Collapse
Affiliation(s)
- Elisa Fernández-Millán
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28040 Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
21
|
Tan Y, Duan T, Li B, Zhang B, Zhu Y, Yan K, Song J, Lv T, Yang J, Jiang L, Yang J, Wen T, Yan L. Sarcopenia defined by psoas muscle index independently predicts long-term survival after living donor liver transplantation in male recipients. Quant Imaging Med Surg 2022; 12:215-228. [PMID: 34993073 DOI: 10.21037/qims-21-314] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/25/2021] [Indexed: 02/05/2023]
Abstract
Background The effect of sarcopenia on long-term outcomes in recipients after living donor liver transplantation (LDLT), including overall survival and hepatocellular carcinoma (HCC) recurrence, remains unclear, especially in China. Methods From 2009 to 2015, 117 adult patients underwent LDLT in our center. In all, 82 patients who had computed tomography images reaching the third lumbar vertebra level within 1 month of LDLT were included; 70 male patients were included in the final analysis after excluding 12 female patients because of poor performance of the calculated cutoff value. Sarcopenia was defined according to the psoas muscle index (PMI) cutoff value, which was calculated based on dynamic time-dependent outcomes using X-tile software. Cox proportional hazards models were used to assess multivariate-adjusted hazards ratios (HRs) to seek potential correlations between sarcopenia and posttransplant outcomes. Results According to the cutoff value of PMI (6.25 cm2/m2), 38 patients (54.3%) were diagnosed with sarcopenia. After an average of 63.3 months of follow-up, 21 patients died after LDLT, 16 in the sarcopenia group and 5 in the non-sarcopenia group, respectively. Sarcopenia was identified as being significantly associated with worse posttransplant overall survival in multivariate analysis, resulting in an HR of 3.22 [95% confidence interval (CI), 1.15-8.98]. Among the 50 recipients with HCC, sarcopenia was significantly associated with HCC recurrence in univariate analysis (HR 2.87, 95% CI, 1.06-7.80) but was not detected as an independent risk factor of HCC recurrence in multivariate analysis, although a trend (tendency)towards significance was observed (HR 2.60, 95% CI, 0.95-7.10; P=0.062). Conclusions Sarcopenia defined by PMI is a feasible and reliable independent predictor of posttransplant overall survival in male LDLT candidates. However, its correlation with posttransplant HCC recurrence remains uncertain.
Collapse
Affiliation(s)
- Yifei Tan
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ting Duan
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Bo Li
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Bohan Zhang
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yunfeng Zhu
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ke Yan
- West China School of Public Health, Sichuan University, Chengdu, China
| | - Jiulin Song
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Lv
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jian Yang
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Li Jiang
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jiayin Yang
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Tianfu Wen
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Lunan Yan
- Liver Transplantation Center, Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Langlois A, Dumond A, Vion J, Pinget M, Bouzakri K. Crosstalk Communications Between Islets Cells and Insulin Target Tissue: The Hidden Face of Iceberg. Front Endocrinol (Lausanne) 2022; 13:836344. [PMID: 35185804 PMCID: PMC8851682 DOI: 10.3389/fendo.2022.836344] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022] Open
Abstract
The regulation of insulin secretion is under control of a complex inter-organ/cells crosstalk involving various metabolites and/or physical connections. In this review, we try to illustrate with current knowledge how β-cells communicate with other cell types and organs in physiological and pathological contexts. Moreover, this review will provide a better understanding of the microenvironment and of the context in which β-cells exist and how this can influence their survival and function. Recent studies showed that β-cell insulin secretion is regulated also by a direct and indirect inter-organ/inter-cellular communication involving various factors, illustrating the idea of "the hidden face of the iceberg". Moreover, any disruption on the physiological communication between β-cells and other cells or organs can participate on diabetes onset. Therefore, for new anti-diabetic treatments' development, it is necessary to consider the entire network of cells and organs involved in the regulation of β-cellular function and no longer just β-cell or pancreatic islet alone. In this context, we discuss here the intra-islet communication, the β-cell/skeletal muscle, β-cell/adipose tissue and β-cell/liver cross talk.
Collapse
|
23
|
Langlois A, Forterre A, Pinget M, Bouzakri K. Impact of moderate exercise on fatty acid oxidation in pancreatic β-cells and skeletal muscle. J Endocrinol Invest 2021; 44:1815-1825. [PMID: 33844166 PMCID: PMC8357749 DOI: 10.1007/s40618-021-01551-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Fatty acids (FA) play a crucial role in glycaemia regulation in healthy and metabolic disorders conditions through various mechanisms. FA oxidation is one of the processes involved in lipid metabolism and can be modulated by exercise. Nowadays, physical activity is known to be an effective strategy for the prevention and treatment of Type 2 Diabetes. Moreover, its intensity, its duration, the sex-gender, the prandial state, exerkines… are as many parameters that can influence glycaemic control. However, the widely debated question is to determine the best type of exercise for patients with metabolic disorders. In this review, we will discuss the impact of exercise intensity, especially moderate activity, on glycaemic control by focussing on FA oxidation in pancreatic β-cells and skeletal muscle. Finally, thanks to all the recent data, we will determine whether moderate physical activity is a good therapeutic strategy and if FA oxidation represents a target of interest to treat diabetic, obese and insulin-resistant patients.
Collapse
Affiliation(s)
- A Langlois
- Centre Européen D'étude du Diabète, Unité Mixte de Recherche de L'Université de Strasbourg « Diabète et Thérapeutique », Strasbourg, France
| | - A Forterre
- Centre Européen D'étude du Diabète, Unité Mixte de Recherche de L'Université de Strasbourg « Diabète et Thérapeutique », Strasbourg, France
| | - M Pinget
- Centre Européen D'étude du Diabète, Unité Mixte de Recherche de L'Université de Strasbourg « Diabète et Thérapeutique », Strasbourg, France
| | - K Bouzakri
- Centre Européen D'étude du Diabète, Unité Mixte de Recherche de L'Université de Strasbourg « Diabète et Thérapeutique », Strasbourg, France.
| |
Collapse
|
24
|
Reed J, Bain S, Kanamarlapudi V. A Review of Current Trends with Type 2 Diabetes Epidemiology, Aetiology, Pathogenesis, Treatments and Future Perspectives. Diabetes Metab Syndr Obes 2021; 14:3567-3602. [PMID: 34413662 PMCID: PMC8369920 DOI: 10.2147/dmso.s319895] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/09/2021] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes (T2D), which has currently become a global pandemic, is a metabolic disease largely characterised by impaired insulin secretion and action. Significant progress has been made in understanding T2D aetiology and pathogenesis, which is discussed in this review. Extrapancreatic pathology is also summarised, which demonstrates the highly multifactorial nature of T2D. Glucagon-like peptide (GLP)-1 is an incretin hormone responsible for augmenting insulin secretion from pancreatic beta-cells during the postprandial period. Given that native GLP-1 has a very short half-life, GLP-1 mimetics with a much longer half-life have been developed, which are currently an effective treatment option for T2D by enhancing insulin secretion in patients. Interestingly, there is continual emerging evidence that these therapies alleviate some of the post-diagnosis complications of T2D. Additionally, these therapies have been shown to induce weight loss in patients, suggesting they could be an alternative to bariatric surgery, a procedure associated with numerous complications. Current GLP-1-based therapies all act as orthosteric agonists for the GLP-1 receptor (GLP-1R). Interestingly, it has emerged that GLP-1R also has allosteric binding sites and agonists have been developed for these sites to test their therapeutic potential. Recent studies have also demonstrated the potential of bi- and tri-agonists, which target multiple hormonal receptors including GLP-1R, to more effectively treat T2D. Improved understanding of T2D aetiology/pathogenesis, coupled with the further elucidation of both GLP-1 activity/targets and GLP-1R mechanisms of activation via different agonists, will likely provide better insight into the therapeutic potential of GLP-1-based therapies to treat T2D.
Collapse
Affiliation(s)
- Josh Reed
- Institute of Life Science 1, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | - Stephen Bain
- Institute of Life Science 1, Medical School, Swansea University, Swansea, SA2 8PP, UK
| | | |
Collapse
|
25
|
Hashida R, Matsuse H, Kawaguchi T, Yoshio S, Bekki M, Iwanaga S, Sugimoto T, Hara K, Koya S, Hirota K, Nakano D, Tsutsumi T, Kanto T, Torimura T, Shiba N. Effects of a low-intensity resistance exercise program on serum miR-630, miR-5703, and Fractalkine/CX3CL1 expressions in subjects with No exercise habits: A preliminary study. Hepatol Res 2021; 51:823-833. [PMID: 34014020 DOI: 10.1111/hepr.13670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/03/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022]
Abstract
AIMS Exercise is effective for the prevention of liver cancer. Exercise exerts biological effects through the regulation of microRNAs (miRNAs) and cytokines/myokines. We aimed to investigate the effects of low-intensity resistance exercise on serum miRNA and cytokine/myokine expressions in subjects with no exercise habits. METHODS We enrolled seven male subjects with no exercise habits in this prospective before-after study. All subjects performed a low-intensity resistance exercise program (three metabolic equivalents, approximately 20 min/session). Serum miRNA expressions were evaluated using microarrays. We performed Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of differentially expressed miRNAs before and after exercise. Serum cytokine/myokine expressions were evaluated using a multiplex panel. RESULTS All subjects completed the exercise program with no adverse events. In the microarray analysis, seven miRNAs showed a significant change between before and after exercise. Of these, microRNA (miR)-630 and miR-5703 showed a >1.5-fold increase (miR-630: 40.7 vs. 69.3 signal intensity, p = 0.0133; miR-5703: 30.7 vs. 55.9 signal intensity, p = 0.0051). KEGG pathway enrichment analysis showed that miR-630- and miR-5703-related genes were enriched in 37 and 5 pathways, including transforming growth factor-beta and Wnt signaling pathways, respectively. In the multiplex analysis, 12 cytokines/myokines showed significant alteration after exercise compared to before exercise. Of these, fractalkine/CX3CL1 showed the most significant up-regulation by exercise (94.5 vs. 109.1 pg/ml, p = 0.0017). CONCLUSIONS A low-intensity resistance exercise program was associated with upregulation of serum miR-630, miR-5703, and fractalkine/CX3CL1 expressions in subjects with no exercise habits. Thus, even low-intensity exercise may alter miRNA and cytokine/myokine expressions in humans.
Collapse
Affiliation(s)
- Ryuki Hashida
- Department of Orthopedics, School of Medicine, Kurume University, Kurume, Japan.,Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Hiroo Matsuse
- Department of Orthopedics, School of Medicine, Kurume University, Kurume, Japan.,Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Sachiyo Yoshio
- Department of Liver Disease, Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Masafumi Bekki
- Department of Orthopedics, School of Medicine, Kurume University, Kurume, Japan.,Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Sohei Iwanaga
- Department of Orthopedics, School of Medicine, Kurume University, Kurume, Japan.,Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Takahiro Sugimoto
- Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Koji Hara
- Department of Orthopedics, School of Medicine, Kurume University, Kurume, Japan.,Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Shunji Koya
- Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Keisuke Hirota
- Division of Rehabilitation, Kurume University Hospital, Kurume, Japan
| | - Dan Nakano
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tsubasa Tsutsumi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuya Kanto
- Department of Liver Disease, Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Naoto Shiba
- Department of Orthopedics, School of Medicine, Kurume University, Kurume, Japan
| |
Collapse
|
26
|
Nakamura S, Yonekura S, Shimosato T, Takaya T. Myogenetic Oligodeoxynucleotide (myoDN) Recovers the Differentiation of Skeletal Muscle Myoblasts Deteriorated by Diabetes Mellitus. Front Physiol 2021; 12:679152. [PMID: 34108889 PMCID: PMC8181739 DOI: 10.3389/fphys.2021.679152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
Skeletal muscle wasting in patients with diabetes mellitus (DM) is a complication of decreased muscle mass and strength, and is a serious risk factor that may result in mortality. Deteriorated differentiation of muscle precursor cells, called myoblasts, in DM patients is considered to be one of the causes of muscle wasting. We recently developed myogenetic oligodeoxynucleotides (myoDNs), which are 18-base single-strand DNAs that promote myoblast differentiation by targeting nucleolin. Herein, we report the applicability of a myoDN, iSN04, to myoblasts isolated from patients with type 1 and type 2 DM. Myogenesis of DM myoblasts was exacerbated concordantly with a delayed shift of myogenic transcription and induction of interleukins. Analogous phenotypes were reproduced in healthy myoblasts cultured with excessive glucose or palmitic acid, mimicking hyperglycemia or hyperlipidemia. iSN04 treatment recovered the deteriorated differentiation of plural DM myoblasts by downregulating myostatin and interleukin-8 (IL-8). iSN04 also ameliorated the impaired myogenic differentiation induced by glucose or palmitic acid. These results demonstrate that myoDNs can directly facilitate myoblast differentiation in DM patients, making them novel candidates for nucleic acid drugs to treat muscle wasting in patients with DM.
Collapse
Affiliation(s)
- Shunichi Nakamura
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Shinichi Yonekura
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Tomohide Takaya
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
27
|
Testa C, DI Lorenzo A, Parlato A, D'Ambrosio G, Merolla A, Pacileo M, Iannuzzo G, Gentile M, Nugara C, Sarullo FM, DE Gregorio C, D'Andrea A, Vigorito C, Venturini E, Giallauria F. Exercise for slowing the progression of atherosclerotic process: effects on inflammatory markers. Panminerva Med 2021; 63:122-132. [PMID: 33565757 DOI: 10.23736/s0031-0808.21.04266-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Atherosclerosis is a dynamic process driven by all cardiovascular risk factors that can be briefly divided into an early and a late phase. Inflammation is one of the fundamental substrates that initiates the atherosclerotic process in the early stages and promotes and maintains it in the final stages. In the last decades, clinical and experimental data have shown that inflammation is supported by mediators that respond to physical activity. The present review aimed at investigating the effect of physical exercise on inflammatory mediators, both the positive ones that have a proinflammatory effect (interleukin 6, c-reactive protein and tumor necrosis factor α, interferon γ, high-mobility group box-1), and the negative ones which have an anti-inflammatory effect (interleukin 10). Pooled data support the evidence that physical exercise can directly modulate the activity of inflammatory cytokines slowing down or preventing the formation of the atherosclerotic stage.
Collapse
Affiliation(s)
- Crescenzo Testa
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Anna DI Lorenzo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Alessandro Parlato
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Giuseppe D'Ambrosio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Aurora Merolla
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Mario Pacileo
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Marco Gentile
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Cinzia Nugara
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Filippo M Sarullo
- Unit of Cardiovascular Rehabilitation, Buccheri La Ferla Fatebenefratelli Hospital, Palermo, Italy
| | - Cesare DE Gregorio
- Unit of Cardiology, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy.,Post-graduate Residency School in Cardiovascular Diseases, Department of Clinical and Experimental Medicine, University Hospital of Messina, Messina, Italy
| | - Antonello D'Andrea
- Unit of Cardiology and Intensive Care, "Umberto I" Hospital, Nocera Inferiore, Salerno, Italy
| | - Carlo Vigorito
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Elio Venturini
- Cardiac Rehabilitation Unit, AUSL Toscana Nord-Ovest, Cecina Civil Hospital, Cecina, Livorno, Italy
| | - Francesco Giallauria
- Department of Translational Medical Sciences, Federico II University, Naples, Italy - .,Faculty of Sciences and Technology, University of New England, Armidale, Australia
| |
Collapse
|
28
|
Impaired capillary tube formation induced by elevated secretion of IL8 involves altered signaling via the CXCR1/PI3K/MMP2 pathway. Mol Biol Rep 2021; 48:601-610. [PMID: 33411234 DOI: 10.1007/s11033-020-06104-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/18/2020] [Indexed: 10/22/2022]
Abstract
Angiogenesis is a multistep process requiring endothelial cell activation, migration, proliferation and tube formation. We recently reported that elevated secretion of interlukin 8 (IL8) by myotubes (MT) from subjects with Type-2 Diabetes (T2D) reduced angiogenesis by human umbilical vein endothelial cells (HUVEC) and human skeletal muscle explants. This lower vascularization was mediated through impaired activation of the phosphatidylinositol 3-kinase (PI3K)-pathway. We sought to investigate additional signaling elements that might mediate reduced angiogenesis. HUVEC were exposed to levels of IL8 equal to those secreted by MT from non-diabetic (ND) and T2D subjects and the involvement of components in the angiogenic response pathway examined. Cellular content of reactive oxygen species and Nitrate secretion were similar after treatment with [ND-IL8] and [T2D-IL8]. CXCR1 protein was down-regulated after treatment with [T2D-IL8] (p < 0.01 vs [ND-IL8] treatment); CXCR2 expression was unaltered. Addition of neutralizing antibodies against CXCR1 and CXCR2 to HUVEC treated with IL8 confirmed that CXCR1 alone mediated the angiogenic response to IL8. A key modulator of angiogenesis is matrix metalloproteinase-2 (MMP2). MMP2 secretion was higher after treatment with [ND-IL8] vs [T2D-IL8] (p < 0.01). MMP2 inhibition reduced tube formation to greater extent with [ND-IL8] than with [T2D-IL8] (p < 0.005). The PI3K-pathway inhibitor LY294002 reduced IL8-induced MMP2 release. IL8 regulation of MMP2 release was CXCR1 dependent, as anti-CXCR1 significantly reduced MMP2 release (p < 0.05). These results suggest that high levels of IL8 secreted by T2D MT trigger reduced capillarization via lower activation of a CXCR1-PI3K pathway, followed by impaired release and activity of MMP2.
Collapse
|
29
|
Razzaque F, Sharif A, Akhtar B, Khan HM, Akhtar MF, Zaib M, Muhammad A, Sohail K, Hamid I, Qaisar N. Tylophora hirsuta (Wall.) Extracts Ameliorate Diabetes Associated with Inflammation in Alloxan-induced Diabetic Rats. Endocr Metab Immune Disord Drug Targets 2021; 21:1031-1042. [PMID: 32955006 DOI: 10.2174/1871530320666200821154340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/03/2020] [Accepted: 07/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Tylophora hirsuta (Wall) has long been used as traditional medicine for the treatment of diabetes. The current study is designed to evaluate the anti-diabetic and anti-inflammatory activity of different extracts of aerial parts of Tylophora hirsuta. METHODS Sequential maceration was conducted to obtain extracts. Total phenolic contents were determined by the Folin-Ciocalteau method. The anti-oxidant activity was assessed by DPPH free radical scavenging assay. The extracts were tested for its inhibitory activity against α-amylase in-vitro. In-vivo anti-diabetic assay was conducted using alloxan-induced diabetic model and OGTT was conducted on normal rats. ELISA was used to determine the pro-inflammatory cytokines (TNF-α and IL-6). The polyphenolic composition of the extract was analyzed using an HPLC system. RESULTS Aqueous extract exhibited highest total phenolic contents (985.24± 3.82 mg GAE/100 g DW), antioxidant activity (IC50 = 786.70 ± 5.23 μg/mL), and alpha-amylase inhibition (IC50 =352.8 μg/mL). The aqueous extract of Tylophora hirsuta showed remarkable in-vivo anti-diabetic activity. Results were compared with standard drug glibenclamide. Alloxan induced diabetic mediated alterations in liver function enzymes, renal function determinants, and lipid parameters were significantly restored in aqueous extract treated diabetic rats. A significant reduction in pro-inflammatory cytokines (p<0.001) was observed when compared to the control group. HPLC analysis confirms the presence of quercetin, gallic acid, cinnamic acid, and p-coumaric acid. CONCLUSION These results showed that Tylophora hirsuta possesses strong anti-diabetic and anti-inflammatory potentials and justify its folklore use for the management of diabetes.
Collapse
Affiliation(s)
- Faisal Razzaque
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Ali Sharif
- Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Jail Road, Lahore, Pakistan
| | - Bushra Akhtar
- Department of Pharmacy, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Humaira M Khan
- Institute of Pharmacy, Faculty of Pharmaceutical and Allied Health Sciences, Lahore College for Women University, Jail Road, Lahore, Pakistan
| | - Muhammad F Akhtar
- Institute of Pharmaceutical Sciences, Ripah International University, Lahore Campus, Lahore, Pakistan
| | - Maryam Zaib
- Johar Institute of Professional Studies, Lahore, Pakistan
| | | | - Kashif Sohail
- Akson College of Health Sciences, Mirpur, Azad Kashmir, Pakistan
| | - Irfan Hamid
- Cadson College of Pharmacy, Kharian, Pakistan
| | - Naeem Qaisar
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| |
Collapse
|
30
|
Faridvand Y, Haddadi P, Nejabati HR, Ghaffari S, Zamani-Gharehchamani E, Nozari S, Nouri M, Jodati A. Sulforaphane modulates CX3CL1/CX3CR1 axis and inflammation in palmitic acid-induced cell injury in C2C12 skeletal muscle cells. Mol Biol Rep 2020; 47:7971-7977. [PMID: 33034881 DOI: 10.1007/s11033-020-05875-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 09/29/2020] [Indexed: 11/28/2022]
Abstract
Studies have shown that sulforaphane (SFN) has potent anti-inflammatory and free radical scavenging effects on obesity and associated disorder such as diabetes, polycystic ovary syndrome, and metabolic syndrome. fractalkine (CX3CL1) and its receptor, CX3CR1, play an important role in muscle metabolism by improving insulin-sensitizing effects. Here, in this study we examined the SFN effect on CX3CL1 and its receptor, CX3CR1, in C2C12 myotubes in palmitic acid (PA)-induced oxidative stress and inflammation. The results showed that PA (750 μM) evoked lipotoxicity as a reduction in cell viability, increased IL-6 and TNF-α expression, and enhanced reactive oxygen species (ROS). However, SFN pretreatment attenuated the levels of, IL-6 and TNF-α in C2C12 myotubes exposure to PA. Moreover, SFN pretreatment up-regulated nuclear factor erythroid related factor 2 (Nrf2) /heme oxygenase-1(HO-1) pathway protein in C2C12 cells as indicated by a decrease in ROS levels. Interestingly, PA also caused an increase in CX3CL1 and CX3CR1 expression that SFN abrogated it. We also found the protective effect of SFN agonist PA-induced lipotoxicity with promotes in UCP3 gene expression in C2C12 cells. Collectively, these findings suggest that SFN hampers the PA-induced inflammation in C2C12 cells by modulation of the Nrf2/HO-1 pathway and CX3CL1/CX3CR1 axis and may propose a new therapeutic approach to protect against obesity-associated disorders in skeletal muscle cells.
Collapse
Affiliation(s)
- Yousef Faridvand
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parinaz Haddadi
- Department of Biochemistry, Faculty of Sciences, Tabriz University, Tabriz, Iran
| | - Hamid Reza Nejabati
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Samira Nozari
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell and Regenerative Medicine (SCARM) Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmadreza Jodati
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
31
|
Chen W, Wang L, You W, Shan T. Myokines mediate the cross talk between skeletal muscle and other organs. J Cell Physiol 2020; 236:2393-2412. [PMID: 32885426 DOI: 10.1002/jcp.30033] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Myokines are muscle-derived cytokines and chemokines that act extensively on organs and exert beneficial metabolic functions in the whole-body through specific signal networks. Myokines as mediators provide the conceptual basis for a whole new paradigm useful for understanding how skeletal muscle communicates with other organs. In this review, we summarize and discuss classes of myokines and their physiological functions in mediating the regulatory roles of skeletal muscle on other organs and the regulation of the whole-body energy metabolism. We review the mechanisms involved in the interaction between skeletal muscle and nonmuscle organs through myokines. Moreover, we clarify the connection between exercise, myokines and disease development, which may contribute to the understanding of a potential mechanism by which physical inactivity affects the process of metabolic diseases via myokines. Based on the current findings, myokines are important factors that mediate the effect of skeletal muscle on other organ functions and whole-body metabolism.
Collapse
Affiliation(s)
- Wentao Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, China.,Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Hangzhou, China
| |
Collapse
|
32
|
Severinsen MCK, Pedersen BK. Muscle-Organ Crosstalk: The Emerging Roles of Myokines. Endocr Rev 2020; 41:5835999. [PMID: 32393961 PMCID: PMC7288608 DOI: 10.1210/endrev/bnaa016] [Citation(s) in RCA: 555] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 05/06/2020] [Indexed: 02/06/2023]
Abstract
Physical activity decreases the risk of a network of diseases, and exercise may be prescribed as medicine for lifestyle-related disorders such as type 2 diabetes, dementia, cardiovascular diseases, and cancer. During the past couple of decades, it has been apparent that skeletal muscle works as an endocrine organ, which can produce and secrete hundreds of myokines that exert their effects in either autocrine, paracrine, or endocrine manners. Recent advances show that skeletal muscle produces myokines in response to exercise, which allow for crosstalk between the muscle and other organs, including brain, adipose tissue, bone, liver, gut, pancreas, vascular bed, and skin, as well as communication within the muscle itself. Although only few myokines have been allocated to a specific function in humans, it has been identified that the biological roles of myokines include effects on, for example, cognition, lipid and glucose metabolism, browning of white fat, bone formation, endothelial cell function, hypertrophy, skin structure, and tumor growth. This suggests that myokines may be useful biomarkers for monitoring exercise prescription for people with, for example, cancer, diabetes, or neurodegenerative diseases.
Collapse
Affiliation(s)
- Mai Charlotte Krogh Severinsen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bente Klarlund Pedersen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Ryan AJ, Ciaraldi TP, Henry RR. Myokine Regulation of Insulin Secretion: Impact of Inflammation and Type 2 Diabetes. Front Physiol 2020; 10:1608. [PMID: 32038288 PMCID: PMC6987462 DOI: 10.3389/fphys.2019.01608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 12/23/2019] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle (SkM) secretes protein factors (myokines) that can exert multiple actions. To study the control of myokine regulation of β-cell function, SkM biopsies were taken from non-diabetic (ND) and Type 2 diabetic (T2D) subjects and satellite cells cultured to myotubes (MT). MT were also treated with lipopolysaccharide (infectious inflammation – II) or a combination of glucose (10 mM), insulin (120 pM), and palmitate (0.4 mM) (metabolic inflammation – MI) to model the inflammatory and metabolic conditions seen in vivo with T2D. Conditioned media (CM) was collected from MT after 24 h and used to treat INS-1 cells for 24 h. Cell viability, total insulin content, glucose-stimulated insulin secretion (GSIS) and maximal (IBMX-stimulated) IS (ISmax) were monitored. Under baseline conditions, CM from ND and T2D MT had no effects on INS-1 cell viability, insulin content, GSIS, or ISmax. After exposure to II, CM from ND-MT augmented GSIS in INS-1 cells by 100 ± 25% over control (p < 0.05); T2D-CM had no effect. After exposure to MI, T2D-CM suppressed GSIS by 35 ± 5% (p < 0.05); ND-CM was without effect. Under either of these conditions cell viability, total insulin content and ISmax were unaffected. Effects of CM on GSIS were lost after CM was boiled. Both augmentation of GSIS by ND-CM from II-treated MT, and suppression by T2D-CM from MI-treated MT, were inhibited by wortmannin, Ro 31-8220, and SB203580. In summary: (1) ND-MT are able to augment GSIS when stressed, (2) T2D-MT responding to a diabetic-like environment secrete myokines that suppress GSIS, (3) Unknown protein factors exert effects specifically on GSIS, possibly through PI-3K, PKC, and/or p38 MAPK. In T2D, both insulin resistance and a suppression of adaptive increased insulin secretion are intrinsic properties of SkM that can contribute to the full T2D phenotype.
Collapse
Affiliation(s)
- Alexander J Ryan
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, LA Jolla, CA, United States
| | - Theodore P Ciaraldi
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, LA Jolla, CA, United States
| | - Robert R Henry
- Veterans Affairs San Diego Healthcare System, San Diego, CA, United States.,Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, LA Jolla, CA, United States
| |
Collapse
|
34
|
Ramírez-Vélez R, Sáez De Asteasu ML, Martínez-Velilla N, Zambon-Ferraresi F, García-Hermoso A, Recarey AE, Fernández-Irigoyen J, Santamaría E, Palomino-Echeverría S, Izquierdo M. Circulating Cytokines and Lower Body Muscle Performance in Older Adults at Hospital Admission. J Nutr Health Aging 2020; 24:1131-1139. [PMID: 33244573 DOI: 10.1007/s12603-020-1480-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Aging-related traits, including gradual loss of skeletal muscle mass and chronic inflammation, are linked to altered body composition and impaired physical functionality, which are important contributing factors to the disabling process. We sought to explore the potential relationship between lower-body muscle strength decline and inflammatory mediators in older adults. METHODS We performed a cross-sectional analysis in 38 older adults admitted to an acute care of the elderly unit (57.9% women, mean age=87.9±4.9 years; mean body mass index [BMI]=26.5±4.7 kg/m2). Clinical and functional outcomes including weight, height, BMI, dependence, physical and cognitive performance, and muscle strength measured by one-repetition maximum (1RM) for leg-extension, leg-press, chest-press and handgrip strength, were assessed. Blood serum content of 59 cytokines, chemokines and growth factors was assessed by protein arrays. Multivariate linear regression analyses were used to examine the relationship between cytokine concentrations and muscle strength parameters. RESULTS After controlling for confounding factors (age, sex, BMI, cumulative illness rating score and physical performance score), 1RM leg-press had a significant negative relationship with GRO (CXCL2) (β= -18.13, p=0.049), MIG (CXCL9) (β= -13.94, p=0.004), IGF-1 (β= -19.63, p=0.003), CK-BETA 8 (CCL23) (β= -28.31, p=0.018) and GCP-2 (CXCL6) (β= -25.78, p=0.004). Likewise, 1RM leg-extension had a significant negative relationship with IGFBP-1 (β= -11.49, p=0.023). CONCLUSIONS Thus, several serum cytokines/chemokines and growth factors are negatively associated with lower muscle strength in older patients. Further investigation is required to elucidate the mechanism of elevated inflammatory mediators leading to lower muscle strength.
Collapse
Affiliation(s)
- R Ramírez-Vélez
- Mikel Izquierdo, PhD, Department of Health Sciences, Public University of Navarra, Av. De Barañain s/n 31008 Pamplona (Navarra) Spain, Tel + 34 948 417876,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Extracellular vesicles (EV), which include exosomes and microvesicles, are membrane-bound particles shed by most cell types and are important mediators of cell-cell communication by delivering their cargo of proteins, miRNA, and mRNA to target cells and altering their function. Here, we provide an overview of what is currently known about EV composition and function in bone and muscle cells and discuss their role in mediating crosstalk between these two tissues as well as their role in musculoskeletal aging. RECENT FINDINGS Recent studies have shown that muscle and bone cells produce EV, whose protein, mRNA, and miRNA cargo reflects the differentiated state of the parental cells. These EV have functional effects within their respective tissues, but evidence is accumulating that they are also shed into the circulation and can have effects on distant tissues. Bone- and muscle-derived EV can alter the differentiation and function of bone and muscle cells. Many of these effects are mediated via small microRNAs that regulate target genes in recipient cells. EV-mediated signaling in muscle and bone is an exciting and emerging field. While considerable progress has been made, much is still to be discovered about the mechanisms regulating EV composition, release, uptake, and function in muscle and bone. A key challenge is to understand more precisely how exosomes function in truly physiological settings.
Collapse
Affiliation(s)
- Weiping Qin
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY, 10468, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri Kansas City, 650 E. 25th Street, Kansas City, MO, 64108, USA.
| |
Collapse
|
36
|
Abstract
Physical activity exerts multiple beneficial effects and the myokine concept provides a framework for understanding the molecular pathways that integrate contracting muscle in the complex network of organ communication. This network includes multiple distinct and distal organs; however, the autocrine and paracrine effects of myokines within skeletal muscle (in which they are produced) also need specific attention. In humans, the functional allocation of myokines remains limited and recent findings on fibre type-specific myokine signatures point to an additional level of complexity. Myokines are involved in the anti-inflammatory effect of physical activity and, therefore, critically counteract insulin resistance and the metabolic perturbations of obesity and type 2 diabetes. Future work needs to address the role of myokines in concert with other crosstalk molecules, and to define their specific impact for metabolic homeostasis.
Collapse
Affiliation(s)
- Jürgen Eckel
- KomIT - Center of Competence for Innovative Diabetes Therapy, German Diabetes Center, Auf'm Hennekamp 65, 40225, Düsseldorf, Germany.
| |
Collapse
|
37
|
Shree N, Venkategowda S, Venkatranganna MV, Datta I, Bhonde RR. Human adipose tissue mesenchymal stem cells as a novel treatment modality for correcting obesity induced metabolic dysregulation. Int J Obes (Lond) 2019; 43:2107-2118. [PMID: 31462691 DOI: 10.1038/s41366-019-0438-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/06/2019] [Accepted: 07/22/2019] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Obesity induced metabolic dysregulation results in cluster of chronic conditions mainly hyperglycemia, hyperinsulinemia, dyslipidemia, diabetes, cardiovascular complications and insulin resistance. To investigate the effect of i.m. injection of human adipose tissue derived mesenchymal stem cells and its secretome in correcting obesity induced metabolic dysregulation in high fat diet fed obese model of mice and understand its mechanism of action. SUBJECTS We injected human adipose tissue derived mesenchymal stem cells (ADMSCs) suspension (CS), conditioned medium (CM) and the cell lysate (CL) intramuscularly in high fat diet (HFD)-induced C57BL/6 mice. Metformin was used as a positive control. ADMSCs were traced in vivo for its bio distribution after injection at different time points. RESULTS ADMSCs-treated mice exhibited remarkable decrease in insulin resistance as quantified by HOMA-IR and triglyceride glucose index with concomitant decrease in oxidized LDL and IL6 as compared with the untreated control. CS injection showed improvement in glucose tolerance and reduction in fatty infiltration in the liver, macrophage infiltration in adipose and hypertrophy of the islets resulting from HFD. Upregulation of miRNA-206, MyoD and increase in protein content of the skeletal muscle in CS-treated mice indicates plausible mechanism of action of ADMSCs treatment in ameliorating IR in HFD mice. CONCLUSION Of all the three treatments, CS was found to be the best. ADMSCs were found to have migrated to different organs in order to bring about the correction in dysregulated metabolism induced by obesity. Our results open up a novel treatment modality for possible therapeutic usage in human subjects by employing autologous or allogeneic ADMSCs for the better management of obesity induced metabolic dysregulation.
Collapse
Affiliation(s)
- Nitya Shree
- School of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, India
| | | | | | | | - Ramesh R Bhonde
- School of Regenerative Medicine, Manipal Academy of Higher Education, Bangalore, India. .,Dr. D.Y. Patil Vidyapeeth, Pune, India.
| |
Collapse
|
38
|
Mizgier ML, Fernández-Verdejo R, Cherfan J, Pinget M, Bouzakri K, Galgani JE. Insights on the Role of Putative Muscle-Derived Factors on Pancreatic Beta Cell Function. Front Physiol 2019; 10:1024. [PMID: 31440170 PMCID: PMC6694406 DOI: 10.3389/fphys.2019.01024] [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: 01/23/2019] [Accepted: 07/24/2019] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscle is a main target of insulin action that plays a pivotal role in postprandial glucose disposal. Importantly, skeletal muscle insulin sensitivity relates inversely with pancreatic insulin secretion, which prompted the hypothesis of the existence of a skeletal muscle-pancreas crosstalk mediated through an endocrine factor. The observation that changes in skeletal muscle glucose metabolism are accompanied by altered insulin secretion supports this hypothesis. Meanwhile, a muscle-derived circulating factor affecting in vivo insulin secretion remains elusive. This factor may correspond to peptides/proteins (so called myokines), exosomes and their cargo, and metabolites. We hereby review the most remarkable evidence encouraging the possibility of such inter-organ communication, with special focus on muscle-derived factors that may potentially mediate such skeletal muscle-pancreas crosstalk.
Collapse
Affiliation(s)
- Maria L Mizgier
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Rodrigo Fernández-Verdejo
- Departamento de Ciencias de la Salud, Nutrición y Dietética, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Julien Cherfan
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Michel Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Karim Bouzakri
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Strasbourg, France
| | - Jose E Galgani
- Departamento de Ciencias de la Salud, Nutrición y Dietética, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Nutrición, Diabetes y Metabolismo, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
39
|
Diaz EC, Børsheim E, Shankar K, Cleves MA, Andres A. Prepregnancy Fat Free Mass and Associations to Glucose Metabolism Before and During Pregnancy. J Clin Endocrinol Metab 2019; 104:1394-1403. [PMID: 30496579 PMCID: PMC7296201 DOI: 10.1210/jc.2018-01381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 11/21/2018] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Our aim was to quantify the individual contribution of prepregnancy (PP) fat-free mass (FFM), expressed as [PP-FFM index (PP-FFMI) = FFM (kg)/height (m2)], on markers of glucose homeostasis before and during pregnancy. METHODS Body composition was assessed in 43 women before pregnancy using air-displacement plethysmography. Blood was drawn at PP and gestational weeks ∼8 and 30. Relationships between body composition (independent) variables and glucose homeostasis (dependent) variables were assessed using adjusted correlations and simple and multiple linear regression analyses. RESULTS PP-FFMI was the strongest predictor of plasma insulin concentration [squared partial correlation (Pr2) = 17, P = 0.007] and homeostasis model assessment of insulin resistance (HOMA2-IR) (Pr2 = 16, P = 0.010). At gestation week 30, PP-FFMI and gestational weight gain (GWG) were the strongest predictors of insulin concentration (PP-FFMI: Pr2 = 20, P = 0.010; GWG: Pr2 = 12, P = 0.052) and HOMA2-IR (PP-FFMI: Pr2 = 19, P = 0.012; GWG: Pr2 = 13, P = 0.045). After accounting for PP fat mass index (PP-FMI), PP-FFMI and GWG were independently associated with first-phase insulin response (PP-FFMI: Pr2 = 20, P = 0.009; GWG: Pr2 = 15, P = 0.025) and second-phase insulin response (PP-FFMI: Pr2 = 19, P = 0.011; GWG: Pr2 = 17, P = 0.016). PP-FMI was the strongest predictor of an oral glucose tolerance test‒derived estimated metabolic clearance rate of glucose (PP-FMI: Pr2 = 14, P = 0.037) and estimated insulin sensitivity index (PP-FMI: Pr2 = 13, P = 0.047). CONCLUSIONS PP-FFMI was a predictor of markers of glucose homeostasis before and during pregnancy. Studies assessing the effect of skeletal muscle quality on metabolic regulation during pregnancy are warranted.
Collapse
Affiliation(s)
- Eva Carolina Diaz
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Correspondence and Reprint Requests: Eva Carolina Diaz, MD, Arkansas Children’s Nutrition Center, 15 Children’s Way, Slot 317, Little Rock, Arkansas 72202. E-mail:
| | - Elisabet Børsheim
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Arkansas Children’s Research Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Kartik Shankar
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Mario Alberto Cleves
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Aline Andres
- Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
40
|
Moresi V, Adamo S, Berghella L. The JAK/STAT Pathway in Skeletal Muscle Pathophysiology. Front Physiol 2019; 10:500. [PMID: 31114509 PMCID: PMC6502894 DOI: 10.3389/fphys.2019.00500] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 04/08/2019] [Indexed: 12/29/2022] Open
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway is a key intracellular mediator of a variety of metabolically relevant hormones and cytokines, including the interleukin-6 (IL-6) family of cytokines. The JAK/STAT pathway transmits extracellular signals to the nucleus, leading to the transcription of genes involved in multiple biological activities. The JAK/STAT pathway has been reported to be required for the homeostasis of different tissues and organs. Indeed, when deregulated, it promotes the initiation and progression of pathological conditions, including cancer, obesity, diabetes, and other metabolic diseases. In skeletal muscle, activation of the JAK/STAT pathway by the IL-6 cytokines accounts for opposite effects: on the one hand, it promotes muscle hypertrophy, by increasing the proliferation of satellite cells; on the other hand, it contributes to muscle wasting. The expression of IL-6 and of key members of the JAK/STAT pathway is regulated at the epigenetic level through histone methylation and histone acetylation mechanisms. Thus, manipulation of the JAK/STAT signaling pathway by specific inhibitors and/or drugs that modulate epigenetics is a promising therapeutic intervention for the treatment of numerous diseases. We focus this review on the JAK/STAT pathway functions in striated muscle pathophysiology and the potential role of IL-6 as an effector of the cross talk between skeletal muscle and other organs.
Collapse
Affiliation(s)
- Viviana Moresi
- Unit of Histology and Medical Embryology, DAHFMO, University La Sapienza, Rome, Italy.,Interuniversity Institute of Myology, Rome, Italy
| | - Sergio Adamo
- Unit of Histology and Medical Embryology, DAHFMO, University La Sapienza, Rome, Italy.,Interuniversity Institute of Myology, Rome, Italy
| | - Libera Berghella
- Unit of Histology and Medical Embryology, DAHFMO, University La Sapienza, Rome, Italy.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
41
|
Garneau L, Aguer C. Role of myokines in the development of skeletal muscle insulin resistance and related metabolic defects in type 2 diabetes. DIABETES & METABOLISM 2019; 45:505-516. [PMID: 30844447 DOI: 10.1016/j.diabet.2019.02.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/04/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
Due to its mass, skeletal muscle is the major site of glucose uptake and an important tissue in the development of type 2 diabetes (T2D). Muscles of patients with T2D are affected with insulin resistance and mitochondrial dysfunction, which result in impaired glucose and fatty acid metabolism. A well-established method of managing the muscle metabolic defects occurring in T2D is physical exercise. During exercise, muscles contract and secrete factors called myokines which can act in an autocrine/paracrine fashion to improve muscle energy metabolism. In patients with T2D, plasma levels as well as muscle levels (mRNA and protein) of some myokines are upregulated, while others are downregulated. The signalling pathways of certain myokines are also altered in skeletal muscle of patients with T2D. Taken together, these findings suggest that myokine secretion is an important factor contributing to the development of muscle metabolic defects during T2D. It is also of interest considering that lack of physical activity is closely linked to the occurrence of this disease. The causal relationships between sedentary behavior, factors secreted by skeletal muscle at rest and during contraction and the development of T2D remain to be elucidated. Many myokines shown to influence muscle energy metabolism still have not been characterized in the context of T2D in skeletal muscle specifically. The purpose of this review is to highlight what is known and what remains to be determined regarding myokine secretion in patients with T2D to uncover potential therapeutic targets for the management of this disease.
Collapse
Affiliation(s)
- L Garneau
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Institut du Savoir Montfort - recherche, Ottawa, ON, K1K 0T2, Canada
| | - C Aguer
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Institut du Savoir Montfort - recherche, Ottawa, ON, K1K 0T2, Canada.
| |
Collapse
|
42
|
Abstract
Physical inactivity is one of the leading health problems in the world. Strong epidemiological and clinical evidence demonstrates that exercise decreases the risk of more than 35 different disorders and that exercise should be prescribed as medicine for many chronic diseases. The physiology and molecular biology of exercise suggests that exercise activates multiple signaling pathways of major health importance. An anti-inflammatory environment is produced with each bout of exercise, and long-term anti-inflammatory effects are mediated via an effect on abdominal adiposity. There is, however, a need to close the gap between knowledge and practice and assure that basic research is translated, implemented, and anchored in society, leading to change of praxis and political statements. In order to make more people move, we need a true translational perspective on exercise as medicine, from molecular and physiological events to infrastructure and architecture, with direct implications for clinical practice and public health.
Collapse
Affiliation(s)
- Bente Klarlund Pedersen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
43
|
Barlow J, Solomon TPJ. Conditioned media from contracting skeletal muscle potentiates insulin secretion and enhances mitochondrial energy metabolism of pancreatic beta-cells. Metabolism 2019; 91:1-9. [PMID: 30445139 DOI: 10.1016/j.metabol.2018.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/16/2018] [Accepted: 11/07/2018] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS In this study, we aimed to examine real-time effects of molecules released by contracting skeletal muscle on the insulin secretory function of β-cells using a novel perifusion platform. We hypothesised that media conditioned by contracting skeletal muscle will influence insulin secretion and mitochondrial energy metabolism in β-cells under normal and type-2 diabetic conditions. METHODS INS-1 832/3 pseudoislets were perifused with media from C2C12 myotubes treated with or without electrical pulse stimulation (EPS; 40 V, 1.0 Hz, 2 ms). Insulin secretory function of pseudoislets was measured before, during, and after EPS to simulate pre-, during-, and post-exercise like effects. Additional experiments were completed in INS-1 832/3 cells under "healthy" and "diabetic-like" conditions as well as human pancreatic islets isolated from nondiabetic and type 2 diabetic donors. RESULTS Insulin secretion increased significantly (P < 0.05) by pseudoislets when perifused with media from myotubes treated with but not without EPS. Conditioned media from EPS-treated myotubes also potentiated insulin secretion from INS-1 832/3 cell monolayers in the presence (P < 0.05) and absence of palmitate (P < 0.001) and in nondiabetic (P < 0.01) and type-2 diabetic (P = 0.06) isolated human islets. Pre-treatment of INS-1 832/3 cells to 24-hour high glucose ± palmitate dampened this effect. Moreover, conditioned media from myotubes treated with EPS significantly increased mitochondrial respiratory activity of INS-1 832/3 cells. CONCLUSION/INTERPRETATION Conditioned media from myotubes treated with EPS potentiates acute insulin release from normal cultured β-cells, nondiabetic islets and Type-2 diabetic islets and is associated with enhanced mitochondrial substrate oxidation.
Collapse
Affiliation(s)
- Jonathan Barlow
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, UK.
| | - Thomas P J Solomon
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, UK; Institute for Metabolism and Systems Research (IMSR), University of Birmingham, UK
| |
Collapse
|
44
|
Rutti S, Dusaulcy R, Hansen JS, Howald C, Dermitzakis ET, Pedersen BK, Pinget M, Plomgaard P, Bouzakri K. Angiogenin and Osteoprotegerin are type II muscle specific myokines protecting pancreatic beta-cells against proinflammatory cytokines. Sci Rep 2018; 8:10072. [PMID: 29968746 PMCID: PMC6030123 DOI: 10.1038/s41598-018-28117-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/13/2018] [Indexed: 12/17/2022] Open
Abstract
Tissue cross-talk is emerging as a determinant way to coordinate the different organs implicated in glucose homeostasis. Among the inter-organ communication factors, muscle-secreted myokines can modulate the function and survival of pancreatic beta-cells. Using primary human myotubes from soleus, vastus lateralis and triceps brachii muscles, we report here that the impact of myokines on beta-cells depends on fiber types and their metabolic status. We show that Type I and type II primary myotubes present specific mRNA and myokine signatures as well as a different sensitivity to TNF-alpha induced insulin resistance. Finally, we show that angiogenin and osteoprotegerin are triceps specific myokines with beta-cell protective actions against proinflammatory cytokines. These results suggest that type I and type II muscles could impact insulin secretion and beta-cell mass differentially in type 2 diabetes through specific myokines secretion.
Collapse
Affiliation(s)
- Sabine Rutti
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200, Strasbourg, France
| | - Rodolphe Dusaulcy
- Molecular Diabetes Laboratory, Division of Endocrinology-Diabetes-Hypertension and Nutrition, University Hospital/University of Geneva Medical School, 1211, Geneva, Switzerland
| | - Jakob S Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Centre of Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Cédric Howald
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1211, Geneva, Switzerland.,Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211, Geneva, Switzerland.,Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, 1211, Geneva, Switzerland.,Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, 1211, Geneva, Switzerland.,Swiss Institute of Bioinformatics, Geneva, Switzerland
| | - Bente K Pedersen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Michel Pinget
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200, Strasbourg, France
| | - Peter Plomgaard
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark.,Centre of Physical Activity Research, Rigshospitalet, Copenhagen, Denmark
| | - Karim Bouzakri
- UMR DIATHEC, EA 7294, Centre Européen d'Etude du Diabète, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Bld René Leriche, 67200, Strasbourg, France.
| |
Collapse
|
45
|
Probing the Effect of Physiological Concentrations of IL-6 on Insulin Secretion by INS-1 832/3 Insulinoma Cells under Diabetic-Like Conditions. Int J Mol Sci 2018; 19:ijms19071924. [PMID: 29966345 PMCID: PMC6073900 DOI: 10.3390/ijms19071924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/18/2018] [Accepted: 06/26/2018] [Indexed: 01/09/2023] Open
Abstract
Exercise improves insulin secretion by pancreatic beta cells (β-cells) in patients with type 2 diabetes, but molecular mechanisms of this effect are yet to be determined. Given that contracting skeletal muscle causes a spike in circulating interleukin-6 (IL-6) levels during exercise, muscle-derived IL-6 is a possible endocrine signal associated with skeletal muscle to β-cell crosstalk. Evidence to support a role of IL-6 in regulating the health and function of β-cells is currently inconsistent and studies investigating the role of IL-6 on the function of β-cells exposed to type 2 diabetic-like conditions are limited and often confounded by supraphysiological IL-6 concentrations. The purpose of this study is to explore the extent by which an exercise-relevant concentration of IL-6 influences the function of pancreatic β-cells exposed to type 2 diabetic-like conditions. Using insulin-secreting INS-1 832/3 cells as an experimental β-cell model, we show that 1-h IL-6 (10 pg/mL) has no effect on insulin secretion under normal conditions and does not restore the loss of insulin secretion caused by elevated glucose ± palmitate or IL-1β. Moreover, treatment of INS-1 832/3 cells to medium collected from C2C12 myotubes conditioned with electrical pulse stimulation does not alter insulin secretion despite significant increases in IL-6. Since insulin secretory defects caused by diabetic-like conditions are neither improved nor worsened by exposure to physiological IL-6 levels, we conclude that the beneficial effect of exercise on β-cell function is unlikely to be driven by muscle-derived IL-6.
Collapse
|
46
|
Barlow JP, Solomon TP. Do skeletal muscle-secreted factors influence the function of pancreatic β-cells? Am J Physiol Endocrinol Metab 2018; 314:E297-E307. [PMID: 29208613 DOI: 10.1152/ajpendo.00353.2017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Skeletal muscle is an endocrine organ that secretes a variety of compounds including proteins (myokines), metabolites, microRNAs (miRNAs), and exosomes, many of which are regulated by exercise and play important roles in endocrine signaling. Interorgan communication via muscle-secreted factors therefore provides a novel area for investigation and implicates the importance of skeletal muscle in the pathophysiology of metabolic diseases such as type 2 diabetes (T2D). Given that underlying molecular mechanisms of T2D are subject of ongoing research, in light of new evidence it is probable that interorgan cross-talk between skeletal muscle and pancreatic β-cells plays an important part. To date, the number of studies published in this field provide the basis of this review. Specifically, we discuss current experimental evidence in support for a role of skeletal muscle to β-cell cross-talk, paying particular attention to muscle-secreted factors including myokines, metabolites, miRNAs, and factors contained within exosomes that influence the function and/or the survival of β-cells in health and disease. In reviewing this evidence, we provide an update on the list of known muscle-secreted factors that have potential to influence the function and/or survival of β-cells under normal and diabetic conditions. We also report limitations of current cross-talk methods and discuss future directions in this growing field.
Collapse
Affiliation(s)
- Jonathan P Barlow
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham , Birmingham, West Midlands , United Kingdom
| | - Thomas P Solomon
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham , Birmingham, West Midlands , United Kingdom
| |
Collapse
|
47
|
Takahashi H, Kotani K, Tanaka K, Egucih Y, Anzai K. Therapeutic Approaches to Nonalcoholic Fatty Liver Disease: Exercise Intervention and Related Mechanisms. Front Endocrinol (Lausanne) 2018; 9:588. [PMID: 30374329 PMCID: PMC6196235 DOI: 10.3389/fendo.2018.00588] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022] Open
Abstract
Exercise training ameliorates nonalcoholic fatty liver disease (NAFLD) as well as obesity and metabolic syndrome. Although it is difficult to eliminate the effects of body weight reduction and increased energy expenditure-some pleiotropic effects of exercise training-a number of studies involving either aerobic exercise training or resistance training programs showed ameliorations in NAFLD that are independent of the improvements in obesity and insulin resistance. In vivo studies have identified effects of exercise training on the liver, which may help to explain the "direct" or "independent" effect of exercise training on NAFLD. Exercise training increases peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) expression, improves mitochondrial function and leads to reduced hepatic steatosis, inflammation, fibrosis, and tumor genesis. Crosstalk between the liver and adipose tissue, skeletal muscle and the microbiome is also a possible mechanism for the effect of exercise training on NAFLD. Although numerous studies have reported benefits of exercise training on NAFLD, the optimal duration and intensity of exercise for the prevention or treatment of NAFLD have not been established. Maintaining adherence of patients with NAFLD to exercise training regimes is another issue to be resolved. The use of comprehensive analytical approaches to identify biomarkers such as hepatokines that specifically reflect the effect of exercise training on liver functions might help to monitor the effect of exercise on NAFLD, and thereby improve adherence of these patients to exercise training. Exercise training is a robust approach for alleviating the pathogenesis of NAFLD, although further clinical and experimental studies are required.
Collapse
Affiliation(s)
- Hirokazu Takahashi
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
- Liver Center, Saga University Hospital, Saga University, Saga, Japan
| | - Kazuhiko Kotani
- Division of Community and Family Medicine, Center for Community Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kenichi Tanaka
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
| | - Yuichiro Egucih
- Liver Center, Saga University Hospital, Saga University, Saga, Japan
| | - Keizo Anzai
- Division of Metabolism and Endocrinology, Faculty of Medicine, Saga University, Saga, Japan
- *Correspondence: Keizo Anzai
| |
Collapse
|
48
|
Chandirasegaran G, Elanchezhiyan C, Ghosh K, Sethupathy S. Berberine chloride ameliorates oxidative stress, inflammation and apoptosis in the pancreas of Streptozotocin induced diabetic rats. Biomed Pharmacother 2017; 95:175-185. [DOI: 10.1016/j.biopha.2017.08.040] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 01/12/2023] Open
|
49
|
Hoffmann C, Weigert C. Skeletal Muscle as an Endocrine Organ: The Role of Myokines in Exercise Adaptations. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a029793. [PMID: 28389517 DOI: 10.1101/cshperspect.a029793] [Citation(s) in RCA: 218] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Exercise stimulates the release of proteins with autocrine, paracrine, or endocrine functions produced in skeletal muscle, termed myokines. Based on the current state of knowledge, the major physiological function of myokines is to protect the functionality and to enhance the exercise capacity of skeletal muscle. Myokines control adaptive processes in skeletal muscle by acting as paracrine regulators of fuel oxidation, hypertrophy, angiogenesis, inflammatory processes, and regulation of the extracellular matrix. Endocrine functions attributed to myokines are involved in body weight regulation, low-grade inflammation, insulin sensitivity, suppression of tumor growth, and improvement of cognitive function. Muscle-derived regulatory RNAs and metabolites, as well as the design of modified myokines, are promising novel directions for treatment of chronic diseases.
Collapse
Affiliation(s)
- Christoph Hoffmann
- Division of Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Cora Weigert
- Division of Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine IV, University Hospital Tübingen, 72076 Tübingen, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München at the University of Tübingen, 72076 Tübingen, Germany.,German Center for Diabetes Research (DZD), 85764 München-Neuherberg, Germany
| |
Collapse
|
50
|
Caton PW, Evans EA, Philpott MP, Hannen RF. Can the skin make you fat? A role for the skin in regulating adipose tissue function and whole-body glucose and lipid homeostasis. Curr Opin Pharmacol 2017; 37:59-64. [PMID: 28985599 DOI: 10.1016/j.coph.2017.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 08/23/2017] [Accepted: 08/29/2017] [Indexed: 01/03/2023]
Abstract
Prevalence of obesity and related complications such as type 2 diabetes (T2D) has increased dramatically in recent decades. Metabolic complications of obesity arise in part due to subcutaneous adipose tissue (SAT) dysfunction. However, it is currently unclear why some obese individuals develop insulin resistance and T2D and others do not. In this review, we discuss the role of the skin in regulating SAT function, and whether presence of inflammatory skin diseases such as psoriasis represent a novel risk mechanism mediating development of obesity-related complications.
Collapse
Affiliation(s)
- Paul W Caton
- Division of Diabetes and Nutritional Sciences, King's College London, London SE1 91UL, UK.
| | - Elizabeth A Evans
- Division of Diabetes and Nutritional Sciences, King's College London, London SE1 91UL, UK
| | - Michael P Philpott
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Rosalind F Hannen
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|