1
|
Aleman J, K R, Wiegand C, Schurdak ME, Vernetti L, Gavlock D, Reese C, DeBiasio R, LaRocca G, Angarita YD, Gough A, Soto-Gutierrez A, Behari J, Yechoor VK, Miedel MT, Stern AM, Banerjee I, Taylor DL. A metabolic dysfunction-associated steatotic liver acinus biomimetic induces pancreatic islet dysfunction in a coupled microphysiology system. Commun Biol 2024; 7:1317. [PMID: 39397070 PMCID: PMC11471816 DOI: 10.1038/s42003-024-07006-7] [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: 05/08/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024] Open
Abstract
Preclinical and clinical studies suggest that lipid-induced hepatic insulin resistance is a primary defect that predisposes to dysfunction in islets, implicating a perturbed liver-pancreas axis underlying the comorbidity of T2DM and MASLD. To investigate this hypothesis, we developed a human biomimetic microphysiological system (MPS) coupling our vascularized liver acinus MPS (vLAMPS) with pancreatic islet MPS (PANIS) enabling MASLD progression and islet dysfunction to be assessed. The modular design of this system (vLAMPS-PANIS) allows intra-organ and inter-organ dysregulation to be deconvoluted. When compared to normal fasting (NF) conditions, under early metabolic syndrome (EMS) conditions, the standalone vLAMPS exhibited characteristics of early stage MASLD, while no significant differences were observed in the standalone PANIS. In contrast, with EMS, the coupled vLAMPS-PANIS exhibited a perturbed islet-specific secretome and a significantly dysregulated glucose stimulated insulin secretion response implicating direct signaling from the dysregulated liver acinus to the islets. Correlations between several pairs of a vLAMPS-derived and a PANIS-derived factors were significantly altered under EMS, as compared to NF conditions, mechanistically connecting MASLD and T2DM associated hepatic-factors with islet-derived GLP-1 synthesis and regulation. Since vLAMPS-PANIS is compatible with patient-specific iPSCs, this platform represents an important step towards addressing patient heterogeneity, identifying disease mechanisms, and advancing precision medicine.
Collapse
Affiliation(s)
- Julio Aleman
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Bioengineering, Pittsburgh, USA
| | - Ravikumar K
- University of Pittsburgh Department of Chemical and Petroleum Engineering, Pittsburgh, USA
| | - Connor Wiegand
- University of Pittsburgh Department of Chemical and Petroleum Engineering, Pittsburgh, USA
| | - Mark E Schurdak
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
| | - Lawrence Vernetti
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
| | - Dillon Gavlock
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | - Celeste Reese
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | - Richard DeBiasio
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | - Greg LaRocca
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
| | | | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA
| | - Alejandro Soto-Gutierrez
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
- University of Pittsburgh Department of Pathology, Pittsburgh, USA
| | - Jaideep Behari
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Vijay K Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, USA
| | - Mark T Miedel
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA
- University of Pittsburgh Liver Research Center, Pittsburgh, USA
- University of Pittsburgh Department of Pathology, Pittsburgh, USA
| | - Andrew M Stern
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA.
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA.
| | - Ipsita Banerjee
- University of Pittsburgh Department of Bioengineering, Pittsburgh, USA.
- University of Pittsburgh Department of Chemical and Petroleum Engineering, Pittsburgh, USA.
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, Pittsburgh, USA.
- University of Pittsburgh Department of Bioengineering, Pittsburgh, USA.
- University of Pittsburgh Department of Computational and Systems Biology, Pittsburgh, USA.
- University of Pittsburgh Liver Research Center, Pittsburgh, USA.
| |
Collapse
|
2
|
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
|
3
|
Nishikawa Y, Sakaguchi H, Takada T, Maeda N, Hyngstrom A. Influence of stimulation frequency on brain-derived neurotrophic factor and cathepsin-B production in healthy young adults. J Comp Physiol B 2024; 194:493-499. [PMID: 38819461 PMCID: PMC11316700 DOI: 10.1007/s00360-024-01566-0] [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: 09/24/2023] [Revised: 04/21/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
Electrical muscle stimulation (EMS) has been shown to stimulate the production of myokines (i.e., brain-derived neurotrophic factor (BDNF)), but the most effective EMS parameters for myokine production have not been fully elucidated. The purpose of this study was to quantify the optimal EMS frequency for stimulating myokine production. This study included sixteen young adults (male, n = 13, age = 27.3 ± 5.5 years). Participants underwent four EMS interventions (20 min each) with the following conditions: (1) 4 Hz, (2) 20 Hz, (3) 80 Hz, and (4) control (no intervention). Blood samples were obtained before and immediately after EMS. For the control condition, blood samples were taken before and after 20 min of quiet sitting. BDNF and cathepsin-B levels were analyzed in serum. Compared to preintervention levels, stimulation at 20 Hz resulted in significantly greater postintervention cathepsin-B and BDNF levels (p < 0.01). On the other hand, the control condition did not result in a significant change between pre- and posttreatment. Furthermore, stimulation at 20 Hz caused significantly larger increases in cathepsin-B and BDNF levels than stimulation at 4-80 Hz or the control condition (p < 0.05). In conclusion, stimulation at 20 Hz effectively causes a robust cathepsin-B and BDNF response. Based on these results, we suggest a new strategy for rehabilitation of people with neurological disorders.
Collapse
Affiliation(s)
- Yuichi Nishikawa
- Faculty of Frontier Engineering, Institute of Science & Engineering, Kanazawa University, Kakuma-machi, Kanazawa, Kanazawa, 920-1192, Japan.
| | - Hiroyuki Sakaguchi
- Graduate School of Frontier Engineering, Kanazawa University, Kanazawa, Japan
| | | | - Noriaki Maeda
- Department of Sport Rehabilitation, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Allison Hyngstrom
- Department of Physical Therapy, Marquette University, Milwaukee, WI, USA
| |
Collapse
|
4
|
Mikołajczyk-Stecyna J, Zuk E, Chmurzynska A, Blatkiewicz M, Jopek K, Rucinski M. The effects of exposure to and timing of a choline-deficient diet during pregnancy and early postnatal life on the skeletal muscle transcriptome of the offspring. Clin Nutr 2024; 43:1503-1515. [PMID: 38729079 DOI: 10.1016/j.clnu.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/09/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is related to muscle loss, but the precise mechanism underlying this association remains unclear. The aim of the present study was thus to determine the influence of maternal fatty liver and dietary choline deficiency during pregnancy and/or lactation periods on the skeletal muscle gene expression profile among 24-day-old male rat offspring. METHODS Histological examination of skeletal muscle tissue specimens obtained from offspring of dams suffering from fatty liver, provided with proper choline intake during pregnancy and lactation (NN), fed a choline-deficient diet during both periods (DD), deprived of choline only during pregnancy (DN), or only during lactation (ND), was performed. The global transcriptome pattern was assessed using a microarray approach (Affymetrix® Rat Gene 2.1 ST Array Strip). The relative expression of selected genes was validated by real-time PCR (qPCR). RESULTS Morphological differences in fat accumulation in skeletal muscle related to choline supply were observed. The global gene expression profile was consistent with abnormal morphological changes. Mettl21c gene was overexpressed in all choline-deficient groups compared to the NN group, while two genes, Cdkn1a and S100a4, were downregulated. Processes of protein biosynthesis were upregulated, and processes related to cell proliferation and lipid metabolism were inhibited in DD, DN, and ND groups compared to the NN group. CONCLUSIONS Prenatal and early postnatal exposure to fatty liver and dietary choline deficiency leads to changes in the transcriptome profile in skeletal muscle of 24-day old male rat offspring and is associated with muscle damage, but the mechanism of it seems to be different at different developmental stages of life. Adequate choline intake during pregnancy and lactation can prevent severe muscle disturbance in the progeny of females suffering from fatty liver.
Collapse
Affiliation(s)
| | - Ewelina Zuk
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Poznań, Poland
| | - Agata Chmurzynska
- Poznań University of Life Sciences, Department of Human Nutrition and Dietetics, Poznań, Poland
| | - Malgorzata Blatkiewicz
- Poznań University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| | - Karol Jopek
- Poznań University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| | - Marcin Rucinski
- Poznań University of Medical Sciences, Department of Histology and Embryology, Poznań, Poland
| |
Collapse
|
5
|
Bozzetti F. Evolving concepts on perioperative nutrition of sarcopenic cancer patients. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:106748. [PMID: 36376142 DOI: 10.1016/j.ejso.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022]
Abstract
The recent recognition of the association of sarcopenia with an increased risk of complications after a surgical procedure calls for rethinking the proper approach of the perioperative care in cancer patients. Sarcopenia is broadly considered in literature according to three different definitions: loss of muscle mass, loss of muscle mass plus reduced muscle function and myosteatosis. The aim of this short review on this issue is to define the excess of risk by type of primary and of surgical procedure, depending on the definition of sarcopenia, to speculate on this association (casual versus causal) and to examine the current therapeutical approaches. The analysis of the data shows that sarcopenia, defined as loss of muscle mass plus reduced muscle function, has the higher predictive power for the occurrence of postoperative complications than the two other definitions, and any definition of sarcopenia works better than the usual indexes or scores of surgical risk. Our analysis supports the concept that: a) sarcopenia is frequently associated with inflammation, but inflammation cannot be considered the only or the absolute cause for sarcopenia, b) sarcopenia is not a simple marker of risk but can have a direct role in the increase of risk. Data on perioperative care of sarcopenic cancer patients are scanty but a correct approach cannot rely on nutritional support alone but on a combined approach of optimized nutrition and exercise, hopefully associated with an anti-inflammatory treatment. This strategy should be applied proactively in keeping with the recent recommendations of the American Society of Clinical Oncology for the medical treatment of advanced cancer patients even if a clear demonstration of effectiveness is still lacking.
Collapse
Affiliation(s)
- Federico Bozzetti
- University of Milan, Faculty of Medicine, via Festa del Perdono, 20100, Milano, Italy.
| |
Collapse
|
6
|
Pinto AC, Tavares P, Neves B, Oliveira PF, Vitorino R, Moreira-Gonçalves D, Ferreira R. Exploiting the therapeutic potential of contracting skeletal muscle-released extracellular vesicles in cancer: Current insights and future directions. J Mol Med (Berl) 2024; 102:617-628. [PMID: 38451309 PMCID: PMC11055777 DOI: 10.1007/s00109-024-02427-7] [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: 09/26/2023] [Revised: 01/09/2024] [Accepted: 02/05/2024] [Indexed: 03/08/2024]
Abstract
The health benefits of exercise training in a cancer setting are increasingly acknowledged; however, the underlying molecular mechanisms remain poorly understood. It has been suggested that extracellular vesicles (EVs) released from contracting skeletal muscles play a key role in mediating the systemic benefits of exercise by transporting bioactive molecules, including myokines. Nevertheless, skeletal muscle-derived vesicles account for only about 5% of plasma EVs, with the immune cells making the largest contribution. Moreover, it remains unclear whether the contribution of skeletal muscle-derived EVs increases after physical exercise or how muscle contraction modulates the secretory activity of other tissues and thus influences the content and profile of circulating EVs. Furthermore, the destination of EVs after exercise is unknown, and it depends on their molecular composition, particularly adhesion proteins. The cargo of EVs is influenced by the training program, with acute training sessions having a greater impact than chronic adaptations. Indeed, there are numerous questions regarding the role of EVs in mediating the effects of exercise, the clarification of which is critical for tailoring exercise training prescriptions and designing exercise mimetics for patients unable to engage in exercise programs. This review critically analyzes the current knowledge on the effects of exercise on the content and molecular composition of circulating EVs and their impact on cancer progression.
Collapse
Affiliation(s)
- Ana Carolina Pinto
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Patrícia Tavares
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and ITR, Translational Research in Population Health, 4200-450, Porto, Portugal
| | - Bruno Neves
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Daniel Moreira-Gonçalves
- CIAFEL, Faculty of Sports, University of Porto and Laboratory for Integrative and ITR, Translational Research in Population Health, 4200-450, Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE and Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
7
|
Aleman J, Ravikumar K, Wiegand C, Schurdak ME, Vernetti L, Gavlock D, Reese C, DeBiasio R, LaRocca G, Angarita YD, Gough A, Soto-Gutierrez A, Behari J, Yechoor V, Miedel MT, Stern AM, Banerjee I, Taylor DL. A metabolic-dysfunction associated steatotic liver acinus biomimetic induces pancreatic islet dysfunction in a coupled microphysiology system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590598. [PMID: 38712135 PMCID: PMC11071380 DOI: 10.1101/2024.04.22.590598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Preclinical and clinical studies suggest that lipid-induced hepatic insulin resistance is a primary defect that predisposes to dysfunction in pancreatic islets, implicating a perturbed liver-pancreas axis underlying the comorbidity of T2DM and MASLD. To investigate this hypothesis, we developed a human biomimetic microphysiological system (MPS) coupling our vascularized liver acinus MPS (vLAMPS) with primary islets on a chip (PANIS) enabling MASLD progression and islet dysfunction to be quantitatively assessed. The modular design of this system (vLAMPS-PANIS) allows intra-organ and inter-organ dysregulation to be deconvoluted. When compared to normal fasting (NF) conditions, under early metabolic syndrome (EMS) conditions, the standalone vLAMPS exhibited characteristics of early stage MASLD, while no significant differences were observed in the standalone PANIS. In contrast, with EMS, the coupled vLAMPS-PANIS exhibited a perturbed islet-specific secretome and a significantly dysregulated glucose stimulated insulin secretion (GSIS) response implicating direct signaling from the dysregulated liver acinus to the islets. Correlations between several pairs of a vLAMPS-derived and a PANIS-derived secreted factors were significantly altered under EMS, as compared to NF conditions, mechanistically connecting MASLD and T2DM associated hepatic factors with islet-derived GLP-1 synthesis and regulation. Since vLAMPS-PANIS is compatible with patient-specific iPSCs, this platform represents an important step towards addressing patient heterogeneity, identifying complex disease mechanisms, and advancing precision medicine.
Collapse
|
8
|
Nielsen SDH, Sahebekhtiari N, Huang Z, Young JF, Rasmussen MK. Comparison of secreted miRNAs and proteins during proliferation and differentiation of bovine satellite cells in culture implies potential roles in regulating myogenesis. Gene 2024; 894:147979. [PMID: 37952749 DOI: 10.1016/j.gene.2023.147979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Cultivated meat is an emerging new technology to produce sustainable meat for the future. The common approach for cultivated meat, is the isolation of satellite cells from donor animals, followed by in vitro proliferation and differentiation into primitive muscle fibers. The transformation of satellite cells into myofibers is tightly orchestrated by intra-cellular signaling, while the inter-cellular signaling is less well understood. Thus, the current study was conducted to map the secretion of potential signaling molecules (MicroRNAs and proteins) during proliferation and differentiation. Primary cultures of satellite cells were grown to 50% and 80% confluence, representing the proliferative phase or serum-starved for 1 and 3 days to induce differentiation. Post incubation in FBS-free media, the media were collected and analyzed for miRNA and protein content using gene-arrays and LC-MS/MS, respectively. When comparing the miRNA secretome at 50% and 80% confluence, we observed four differentially expressed miRNA, while only five were differentially expressed when comparing Day 1 to Day 3. A subsequent in silico analysis suggested that pathways of importance for myogenesis, e.g., MAPK and AMPK signaling, could be regulated by the secreted miRNAs. In addition, >300 proteins were secreted, including insulin-like growth factor 1 binding proteins 2, 3, 4, 5 and 6. In conclusion, this study demonstrated differential secretion of several miRNAs and proteins during both proliferation and differentiation of bovine satellite cells in vitro.
Collapse
Affiliation(s)
| | - Navid Sahebekhtiari
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | - Ziyu Huang
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | - Jette Feveile Young
- Department of Food Science, Aarhus University, Agro Food Park 48, 8200 Aarhus N, Denmark
| | | |
Collapse
|
9
|
Dowling P, Trollet C, Negroni E, Swandulla D, Ohlendieck K. How Can Proteomics Help to Elucidate the Pathophysiological Crosstalk in Muscular Dystrophy and Associated Multi-System Dysfunction? Proteomes 2024; 12:4. [PMID: 38250815 PMCID: PMC10801633 DOI: 10.3390/proteomes12010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
This perspective article is concerned with the question of how proteomics, which is a core technique of systems biology that is deeply embedded in the multi-omics field of modern bioresearch, can help us better understand the molecular pathogenesis of complex diseases. As an illustrative example of a monogenetic disorder that primarily affects the neuromuscular system but is characterized by a plethora of multi-system pathophysiological alterations, the muscle-wasting disease Duchenne muscular dystrophy was examined. Recent achievements in the field of dystrophinopathy research are described with special reference to the proteome-wide complexity of neuromuscular changes and body-wide alterations/adaptations. Based on a description of the current applications of top-down versus bottom-up proteomic approaches and their technical challenges, future systems biological approaches are outlined. The envisaged holistic and integromic bioanalysis would encompass the integration of diverse omics-type studies including inter- and intra-proteomics as the core disciplines for systematic protein evaluations, with sophisticated biomolecular analyses, including physiology, molecular biology, biochemistry and histochemistry. Integrated proteomic findings promise to be instrumental in improving our detailed knowledge of pathogenic mechanisms and multi-system dysfunction, widening the available biomarker signature of dystrophinopathy for improved diagnostic/prognostic procedures, and advancing the identification of novel therapeutic targets to treat Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Capucine Trollet
- Center for Research in Myology U974, Sorbonne Université, INSERM, Myology Institute, 75013 Paris, France; (C.T.); (E.N.)
| | - Elisa Negroni
- Center for Research in Myology U974, Sorbonne Université, INSERM, Myology Institute, 75013 Paris, France; (C.T.); (E.N.)
| | - Dieter Swandulla
- Institute of Physiology, Faculty of Medicine, University of Bonn, D53115 Bonn, Germany;
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
10
|
Dowling P, Swandulla D, Ohlendieck K. Mass Spectrometry-Based Proteomic Technology and Its Application to Study Skeletal Muscle Cell Biology. Cells 2023; 12:2560. [PMID: 37947638 PMCID: PMC10649384 DOI: 10.3390/cells12212560] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Voluntary striated muscles are characterized by a highly complex and dynamic proteome that efficiently adapts to changed physiological demands or alters considerably during pathophysiological dysfunction. The skeletal muscle proteome has been extensively studied in relation to myogenesis, fiber type specification, muscle transitions, the effects of physical exercise, disuse atrophy, neuromuscular disorders, muscle co-morbidities and sarcopenia of old age. Since muscle tissue accounts for approximately 40% of body mass in humans, alterations in the skeletal muscle proteome have considerable influence on whole-body physiology. This review outlines the main bioanalytical avenues taken in the proteomic characterization of skeletal muscle tissues, including top-down proteomics focusing on the characterization of intact proteoforms and their post-translational modifications, bottom-up proteomics, which is a peptide-centric method concerned with the large-scale detection of proteins in complex mixtures, and subproteomics that examines the protein composition of distinct subcellular fractions. Mass spectrometric studies over the last two decades have decisively improved our general cell biological understanding of protein diversity and the heterogeneous composition of individual myofibers in skeletal muscles. This detailed proteomic knowledge can now be integrated with findings from other omics-type methodologies to establish a systems biological view of skeletal muscle function.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Dieter Swandulla
- Institute of Physiology, Faculty of Medicine, University of Bonn, D53115 Bonn, Germany;
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland;
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
11
|
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
|
12
|
Wight TN, Day AJ, Kang I, Harten IA, Kaber G, Briggs DC, Braun KR, Lemire JM, Kinsella MG, Hinek A, Merrilees MJ. V3: an enigmatic isoform of the proteoglycan versican. Am J Physiol Cell Physiol 2023; 325:C519-C537. [PMID: 37399500 PMCID: PMC10511178 DOI: 10.1152/ajpcell.00059.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 07/05/2023]
Abstract
V3 is an isoform of the extracellular matrix (ECM) proteoglycan (PG) versican generated through alternative splicing of the versican gene such that the two major exons coding for sequences in the protein core that support chondroitin sulfate (CS) glycosaminoglycan (GAG) chain attachment are excluded. Thus, versican V3 isoform carries no GAGs. A survey of PubMed reveals only 50 publications specifically on V3 versican, so it is a very understudied member of the versican family, partly because to date there are no antibodies that can distinguish V3 from the CS-carrying isoforms of versican, that is, to facilitate functional and mechanistic studies. However, a number of in vitro and in vivo studies have identified the expression of the V3 transcript during different phases of development and in disease, and selective overexpression of V3 has shown dramatic phenotypic effects in "gain and loss of function" studies in experimental models. Thus, we thought it would be useful and instructive to discuss the discovery, characterization, and the putative biological importance of the enigmatic V3 isoform of versican.
Collapse
Affiliation(s)
- Thomas N Wight
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States
| | - Anthony J Day
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, United Kingdom
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States
| | - Ingrid A Harten
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States
| | - Gernot Kaber
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States
| | - David C Briggs
- Signalling and Structural Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Kathleen R Braun
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States
| | - Joan M Lemire
- Department of Biology, Tufts University, Medford, Massachusetts, United States
| | - Michael G Kinsella
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, United States
| | - Aleksander Hinek
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mervyn J Merrilees
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| |
Collapse
|
13
|
Vints WAJ, Gökçe E, Langeard A, Pavlova I, Çevik ÖS, Ziaaldini MM, Todri J, Lena O, Sakkas GK, Jak S, Zorba (Zormpa) I, Karatzaferi C, Levin O, Masiulis N, Netz Y. Myokines as mediators of exercise-induced cognitive changes in older adults: protocol for a comprehensive living systematic review and meta-analysis. Front Aging Neurosci 2023; 15:1213057. [PMID: 37520128 PMCID: PMC10374322 DOI: 10.3389/fnagi.2023.1213057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/20/2023] [Indexed: 08/01/2023] Open
Abstract
Background The world's population is aging, but life expectancy has risen more than healthy life expectancy (HALE). With respect to brain and cognition, the prevalence of neurodegenerative disorders increases with age, affecting health and quality of life, and imposing significant healthcare costs. Although the effects of physical exercise on cognition in advanced age have been widely explored, in-depth fundamental knowledge of the underlying mechanisms of the exercise-induced cognitive improvements is lacking. Recent research suggests that myokines, factors released into the blood circulation by contracting skeletal muscle, may play a role in mediating the beneficial effect of exercise on cognition. Our goal in this ongoing (living) review is to continuously map the rapidly accumulating knowledge on pathways between acute or chronic exercise-induced myokines and cognitive domains enhanced by exercise. Method Randomized controlled studies will be systematically collected at baseline and every 6 months for at least 5 years. Literature search will be performed online in PubMed, EMBASE, PsycINFO, Web of Science, SportDiscus, LILACS, IBECS, CINAHL, SCOPUS, ICTRP, and ClinicalTrials.gov. Risk of bias will be assessed using the Revised Cochrane Risk of Bias tool (ROB 2). A random effects meta-analysis with mediation analysis using meta-analytic structural equation modeling (MASEM) will be performed. The primary research question is to what extent exercise-induced myokines serve as mediators of cognitive function. Secondarily, the pooled effect size of specific exercise characteristics (e.g., mode of exercise) or specific older adults' populations (e.g., cognitively impaired) on the relationship between exercise, myokines, and cognition will be assessed. The review protocol was registered in PROSPERO (CRD42023416996). Discussion Understanding the triad relationship between exercise, myokines and cognition will expand the knowledge on multiple integrated network systems communicating between skeletal muscles and other organs such as the brain, thus mediating the beneficial effects of exercise on health and performance. It may also have practical implications, e.g., if a certain myokine is found to be a mediator between exercise and cognition, the optimal exercise characteristics for inducing this myokine can be prescribed. The living review is expected to improve our state of knowledge and refine exercise regimes for enhancing cognitive functioning in diverse older adults' populations. Registration Systematic review and meta-analysis protocol was registered with the International Prospective Register of Systematic Reviews (PROSPERO) on the 24th of April 2023 (registration number CRD42023416996).
Collapse
Affiliation(s)
- Wouter A. J. Vints
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- Department of Rehabilitation Medicine, Research School Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, Netherlands
- Adelante Zorggroep Centre of Expertise in Rehabilitation and Audiology, Hoensbroek, Netherlands
| | - Evrim Gökçe
- Sports Rehabilitation Laboratory, Ankara City Hospital, Ankara, Türkiye
| | | | - Iuliia Pavlova
- Department of Theory and Methods of Physical Culture, Lviv State University of Physical Culture, Lviv, Ukraine
| | | | | | - Jasemin Todri
- Department of Physiotherapy, Universidad Catolica San Antonio (UCAM), Murcia, Spain
| | - Orges Lena
- Department of Physiotherapy, Universidad Catolica San Antonio (UCAM), Murcia, Spain
| | - Giorgos K. Sakkas
- Lifestyle Medicine and Experimental Physiology and Myology Lab, Department of Physical Education and Sports Science, The Center of Research and Evaluation of Human Performance (CREHP), University of Thessaly, National and Kapodistrian University of Athens (TEFAA) Campus, Karyes, Greece
| | - Suzanne Jak
- Research Institute of Child Development and Education, University of Amsterdam, Amsterdam, Netherlands
| | | | - Christina Karatzaferi
- Lifestyle Medicine and Experimental Physiology and Myology Lab, Department of Physical Education and Sports Science, The Center of Research and Evaluation of Human Performance (CREHP), University of Thessaly, National and Kapodistrian University of Athens (TEFAA) Campus, Karyes, Greece
| | - Oron Levin
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- Movement Control and Neuroplasticity Research Group, Group Biomedical Sciences, Catholic University of Leuven, Heverlee, Belgium
| | - Nerijus Masiulis
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
| | - Yael Netz
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
- The Levinsky-Wingate Academic Center, Wingate Campus, Netanya, Israel
| |
Collapse
|
14
|
Arjunan A, Song J. Pharmacological and physiological roles of adipokines and myokines in metabolic-related dementia. Biomed Pharmacother 2023; 163:114847. [PMID: 37150030 DOI: 10.1016/j.biopha.2023.114847] [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: 03/21/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023] Open
Abstract
Dementia is a detrimental neuropathologic condition with considerable physical, mental, social, and financial impact on patients and society. Patients with metabolic syndrome (MetS), a group of diseases that occur in tandem and increase the risk of neurologic diseases, have a higher risk of dementia. The ratio between muscle and adipose tissue is crucial in MetS, as these contain many hormones, including myokines and adipokines, which are involved in crosstalk and local paracrine/autocrine interactions. Evidence suggests that abnormal adipokine and myokine synthesis and release may be implicated in various MetS, such as atherosclerosis, diabetic mellitus (DM), and dyslipidemia, but their precise role is unclear. Here we review the literature on adipokine and myokine involvement in MetS-induced dementia via glucose and insulin homeostasis regulation, neuroinflammation, vascular dysfunction, emotional changes, and cognitive function.
Collapse
Affiliation(s)
- Archana Arjunan
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Seoyangro 264, Hwasun 58128, Republic of Korea.
| |
Collapse
|
15
|
Kuramoto K, Liang H, Hong JH, He C. Exercise-activated hepatic autophagy via the FN1-α5β1 integrin pathway drives metabolic benefits of exercise. Cell Metab 2023; 35:620-632.e5. [PMID: 36812915 PMCID: PMC10079584 DOI: 10.1016/j.cmet.2023.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/01/2022] [Accepted: 01/26/2023] [Indexed: 02/24/2023]
Abstract
How exercise elicits systemic metabolic benefits in both muscles and non-contractile tissues is unclear. Autophagy is a stress-induced lysosomal degradation pathway that mediates protein and organelle turnover and metabolic adaptation. Exercise activates autophagy in not only contracting muscles but also non-contractile tissues including the liver. However, the role and mechanism of exercise-activated autophagy in non-contractile tissues remain mysterious. Here, we show that hepatic autophagy activation is essential for exercise-induced metabolic benefits. Plasma or serum from exercised mice is sufficient to activate autophagy in cells. By proteomic studies, we identify fibronectin (FN1), which was previously considered as an extracellular matrix protein, as an exercise-induced, muscle-secreted, autophagy-inducing circulating factor. Muscle-secreted FN1 mediates exercise-induced hepatic autophagy and systemic insulin sensitization via the hepatic receptor α5β1 integrin and the downstream IKKα/β-JNK1-BECN1 pathway. Thus, we demonstrate that hepatic autophagy activation drives exercise-induced metabolic benefits against diabetes via muscle-secreted soluble FN1 and hepatic α5β1 integrin signaling.
Collapse
Affiliation(s)
- Kenta Kuramoto
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Huijia Liang
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jung-Hwa Hong
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Congcong He
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
16
|
Feng Y, Cui Z, Lu X, Gong H, Liu X, Wang H, Cheng H, Gao H, Shi X, Li Y, Ye H, Zhang Q, Kong X. Transcriptomics Dissection of Calorie Restriction and Exercise Training in Brown Adipose Tissue and Skeletal Muscle. Nutrients 2023; 15:nu15041047. [PMID: 36839405 PMCID: PMC9966723 DOI: 10.3390/nu15041047] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Calorie restriction (CR) and exercise training (EX) are two critical lifestyle interventions for the prevention and treatment of metabolic diseases, such as obesity and diabetes. Brown adipose tissue (BAT) and skeletal muscle are two important organs for the generation of heat. Here, we undertook detailed transcriptional profiling of these two thermogenic tissues from mice treated subjected to CR and/or EX. We found transcriptional reprogramming of BAT and skeletal muscle as a result of CR but little from EX. Consistent with this, CR induced alterations in the expression of genes encoding adipokines and myokines in BAT and skeletal muscle, respectively. Deconvolution analysis showed differences in the subpopulations of myogenic cells, mesothelial cells and endogenic cells in BAT and in the subpopulations of satellite cells, immune cells and endothelial cells in skeletal muscle as a result of CR or EX. NicheNet analysis, exploring potential inter-organ communication, indicated that BAT and skeletal muscle could mutually regulate their fatty acid metabolism and thermogenesis through ligands and receptors. These data comprise an extensive resource for the study of thermogenic tissue molecular responses to CR and/or EX in a healthy state.
Collapse
Affiliation(s)
- Yonghao Feng
- Department of Endocrinology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Zhicheng Cui
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaodan Lu
- Precision Medicine Center, Jilin Province General Hospital, Changchun 130021, China
| | - Hongyu Gong
- School of Life Sciences, Inner Mongolia University, Hohhot 010000, China
| | - Xiaoyu Liu
- School of Life Sciences, Inner Mongolia University, Hohhot 010000, China
| | - Hui Wang
- Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Haoyu Cheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Huanqing Gao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaohong Shi
- Department of Endocrinology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Yiming Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hongying Ye
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qiongyue Zhang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai 200040, China
- Correspondence: (Q.Z.); (X.K.)
| | - Xingxing Kong
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Huashan Hospital, School of Life Sciences, Fudan University, Shanghai 200438, China
- Correspondence: (Q.Z.); (X.K.)
| |
Collapse
|
17
|
Nishikawa Y, Sakaguchi H, Kawade S, Maeda N, Tanaka S, Hyngstrom A. Electrical muscle stimulation in young adults: effect of muscle volume on brain-derived neurotrophic factor levels. Eur J Appl Physiol 2023; 123:361-366. [PMID: 36301337 DOI: 10.1007/s00421-022-05078-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/18/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE Electrical muscle stimulation (EMS) is known to be effective at stimulating brain-derived neurotrophic factor (BDNF) levels, but the relationship between the volume of muscle stimulated and BDNF levels is not clear. The purpose of this study was to quantify BDNF as a function of muscle volume stimulated in young adults. METHODS Twelve young adults (male, n = 9, age = 27.3 ± 5.5 years) were enrolled in this study. Participants completed three testing conditions in randomized order: 23 min of maximum tolerated bilateral stimulation of (1) the quadriceps muscle or (2) the musculature of the entire lower limbs and (3) control testing and retesting after 23 min without an intervention. Blood samples were collected before, immediately after, 20 min after, and 40 min after the intervention when EMS was applied to the thighs or the entire lower limb conditions. Serum obtained from blood collection was used for BDNF analysis. RESULTS The delta value of BDNF for the test and retest in the control condition was - 42.1 ± 73.8 pg/mL, and there was no significant difference between the test and retest BDNF. Compared to stimulation of the quadriceps muscle, stimulation of the entire lower limbs produced significantly higher BDNF at 20 min post-treatment than those at pre-treatment or 40 min post-treatment, and BDNF was also significantly higher immediately post-treatment than those at pre-treatment. Only stimulation of the quadriceps muscle did not induce a significant change between pre- and post-treatment. CONCLUSION Our findings suggest that the volume of muscle stimulation is important for increased BDNF.
Collapse
Affiliation(s)
- Yuichi Nishikawa
- Faculty of Frontier Engineering, Institute of Science & Engineering, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan.
| | - Hiroyuki Sakaguchi
- Graduate School of Frontier Engineering, Kanazawa University, Kanazawa, Japan
| | | | - Noriaki Maeda
- Division of Sports Rehabilitation, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinobu Tanaka
- Faculty of Frontier Engineering, Institute of Science & Engineering, Kanazawa University, Kakuma-Machi, Kanazawa, 920-1192, Japan
| | - Allison Hyngstrom
- Department of Physical Therapy, Marquette University, Milwaukee, WI, USA
| |
Collapse
|
18
|
Joo SK, Kim W. Interaction between sarcopenia and nonalcoholic fatty liver disease. Clin Mol Hepatol 2023; 29:S68-S78. [PMID: 36472051 PMCID: PMC10029947 DOI: 10.3350/cmh.2022.0358] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Sarcopenia and nonalcoholic fatty liver disease (NAFLD) are common health problems related to aging. Despite the differences in their diagnostic methods, several cross-sectional and longitudinal studies have revealed the close link between sarcopenia and NAFLD. Sarcopenia and NAFLD are linked by several shared pathogenetic mechanisms, including insulin resistance, hormonal imbalance, systemic inflammation, myostatin and adiponectin dysregulation, nutritional deficiencies, and physical inactivity, thus implicating a bidirectional relationship between sarcopenia and NAFLD. However, there is not sufficient data to support a direct causal relationship between sarcopenia and NAFLD. Moreover, it is currently difficult to conclude whether sarcopenia is a risk factor for nonalcoholic steatohepatitis (NASH) or is a consequence of NASH. Therefore, this review intends to touch on the shared common mechanisms and the bidirectional relationship between sarcopenia and NAFLD.
Collapse
Affiliation(s)
- Sae Kyung Joo
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Won Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
19
|
Association between Regional Body Muscle Mass and Non-Alcoholic Fatty Liver Disease: An Observational Study Using Data from the REACTION Study. J Pers Med 2023; 13:jpm13020209. [PMID: 36836444 PMCID: PMC9959461 DOI: 10.3390/jpm13020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/27/2023] Open
Abstract
Background and aims: Regional muscle distribution is associated with abdominal obesity and metabolic syndrome. However, the relationship between muscle distribution and nonalcoholic fatty liver disease (NAFLD) remains unclear. This study was to determine the relationship between regional muscle distribution and the risk and severity of NAFLD. Methods: This cross-sectional study ultimately included 3161 participants. NAFLD diagnosed by ultrasonography was classified into three groups (non, mild, and moderate/severe). We estimated the regional body muscle mass (lower limbs, upper limbs, extremities, and trunk) through multifrequency bioelectrical impedance analysis (BIA). The relative muscle mass was defined as the muscle mass adjusted for the body mass index (BMI). Results: NAFLD participants accounted for 29.9% (945) of the study's population. Individuals with a higher lower limb, extremity, and trunk muscle mass had a lower risk of NAFLD (p < 0.001). Patients with moderate/severe NAFLD had a lower muscle mass of the lower limbs and trunk than patients with mild NAFLD (p < 0.001), while the muscle mass of the upper limbs and extremities did not differ significantly between the two groups. Moreover, similar results were found for both sexes and among different age groups. Conclusions: A higher muscle mass of the lower limbs, extremities, and trunk was negatively associated with the risk of NAFLD. A lower muscle mass of the limbs and trunk was inversely associated with the severity of NAFLD. This study provides a new theoretical basis for the development of individualized exercise prescriptions for the prevention of NAFLD in non-NAFLD patients.
Collapse
|
20
|
Arabzadeh E, Ghassemi Gil-kalayeh Z, Gholami M, Abed Natanzi H, Ebrahimi S. The effect of 8 weeks of circuit resistance training on serum levels of decorin and IGF-I in sedentary young men. SPORT SCIENCES FOR HEALTH 2023. [DOI: 10.1007/s11332-022-01035-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
21
|
Aas V, Øvstebø R, Brusletto BS, Aspelin T, Trøseid AMS, Qureshi S, Eid DSO, Olstad OK, Nyman TA, Haug KBF. Distinct microRNA and protein profiles of extracellular vesicles secreted from myotubes from morbidly obese donors with type 2 diabetes in response to electrical pulse stimulation. Front Physiol 2023; 14:1143966. [PMID: 37064893 PMCID: PMC10098097 DOI: 10.3389/fphys.2023.1143966] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/09/2023] [Indexed: 04/18/2023] Open
Abstract
Lifestyle disorders like obesity, type 2 diabetes (T2D), and cardiovascular diseases can be prevented and treated by regular physical activity. During exercise, skeletal muscles release signaling factors that communicate with other organs and mediate beneficial effects of exercise. These factors include myokines, metabolites, and extracellular vesicles (EVs). In the present study, we have examined how electrical pulse stimulation (EPS) of myotubes, a model of exercise, affects the cargo of released EVs. Chronic low frequency EPS was applied for 24 h to human myotubes isolated and differentiated from biopsy samples from six morbidly obese females with T2D, and EVs, both exosomes and microvesicles (MV), were isolated from cell media 24 h thereafter. Size and concentration of EV subtypes were characterized by nanoparticle tracking analysis, surface markers were examined by flow cytometry and Western blotting, and morphology was confirmed by transmission electron microscopy. Protein content was assessed by high-resolution proteomic analysis (LC-MS/MS), non-coding RNA was quantified by Affymetrix microarray, and selected microRNAs (miRs) validated by real time RT-qPCR. The size and concentration of exosomes and MV were unaffected by EPS. Of the 400 miRs identified in the EVs, EPS significantly changed the level of 15 exosome miRs, of which miR-1233-5p showed the highest fold change. The miR pattern of MV was unaffected by EPS. Totally, about 1000 proteins were identified in exosomes and 2000 in MV. EPS changed the content of 73 proteins in exosomes, 97 in MVs, and of these four were changed in both exosomes and MV (GANAB, HSPA9, CNDP2, and ATP5B). By matching the EPS-changed miRs and proteins in exosomes, 31 targets were identified, and among these several promising signaling factors. Of particular interest were CNDP2, an enzyme that generates the appetite regulatory metabolite Lac-Phe, and miR-4433b-3p, which targets CNDP2. Several of the regulated miRs, such as miR-92b-5p, miR-320b, and miR-1233-5p might also mediate interesting signaling functions. In conclusion, we have used a combined transcriptome-proteome approach to describe how EPS affected the cargo of EVs derived from myotubes from morbidly obese patients with T2D, and revealed several new factors, both miRs and proteins, that might act as exercise factors.
Collapse
Affiliation(s)
- Vigdis Aas
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway
- *Correspondence: Vigdis Aas, ; Kari Bente Foss Haug,
| | - Reidun Øvstebø
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | | | - Trude Aspelin
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
| | | | - Saba Qureshi
- Department of Life Sciences and Health, Oslo Metropolitan University (OsloMet), Oslo, Norway
| | | | | | - Tuula A. Nyman
- Department of Immunology, University of Oslo, and Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kari Bente Foss Haug
- Department of Medical Biochemistry, Oslo University Hospital, Ullevål, Oslo, Norway
- *Correspondence: Vigdis Aas, ; Kari Bente Foss Haug,
| |
Collapse
|
22
|
“Ferrocrinology”—Iron Is an Important Factor Involved in Gluco- and Lipocrinology. Nutrients 2022; 14:nu14214693. [DOI: 10.3390/nu14214693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/04/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
“Ferrocrinology” is the term used to describe the study of iron effects on the functioning of adipose tissue, which together with muscle tissue makes the largest endocrine organ in the human body. By impairing exercise capacity, reducing AMP-activated kinase activity, and enhancing insulin resistance, iron deficiency can lead to the development of obesity and type 2 diabetes mellitus. Due to impaired browning of white adipose tissue and reduced mitochondrial iron content in adipocytes, iron deficiency (ID) can cause dysfunction of brown adipose tissue. By reducing ketogenesis, aconitase activity, and total mitochondrial capacity, ID impairs muscle performance. Another important aspect is the effect of ID on the impairment of thermogenesis due to reduced binding of thyroid hormones to their nuclear receptors, with subsequently impaired utilization of norepinephrine in tissues, and impaired synthesis and distribution of cortisol, which all make the body’s reactivity to stress in ID more pronounced. Iron deficiency can lead to the development of the most common endocrinopathy, autoimmune thyroid disease. In this paper, we have discussed the role of iron in the cross-talk between glucocrinology, lipocrinology and myocrinology, with thyroid hormones acting as an active bystander.
Collapse
|
23
|
Liu H, Wu B, Shi X, Cao Y, Zhao X, Liang D, Qin Q, Liang X, Lu W, Wang D, Liu J. Aerobic exercise-induced circulating extracellular vesicle combined decellularized dermal matrix hydrogel facilitates diabetic wound healing by promoting angiogenesis. Front Bioeng Biotechnol 2022; 10:903779. [PMID: 36082169 PMCID: PMC9445842 DOI: 10.3389/fbioe.2022.903779] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Insufficient blood supply results in unsatisfactory wound healing, especially for challenging wound repair such as diabetic wound defects. Regular exercise training brings a lot of benefits to cardiovascular fitness and metabolic health including attenuation of T2DM progression. Circulating extracellular vesicles (EVs) are postulated to carry a variety of signals involved in tissue crosstalk by their modified cargoes, representing novel mechanisms for the effects of exercise. Prominently, both acute and chronic aerobic exercise training can promote the release of exercise-induced cytokines and enhance the angiogenic function of circulating angiogenic cell–derived EVs.Methods: We investigated the possible angiogenesis potential of aerobic exercise-induced circulating EVs (EXE-EVs) on diabetic wound healing. Circulating EVs were isolated from the plasma of rats subjected to 4 weeks of moderate aerobic exercise or sedentariness 24 h after the last training session. The therapeutic effect of circulating EVs was evaluated in vitro by proliferation, migration, and tube formation assays of human umbilical vein endothelial cells (HUVECs), as well as in vivo by quantification of angiogenesis and cutaneous wound healing in diabetic rats.Results: The number of circulating EVs did not change significantly in exercised rats 24 h post-exercise in comparison with the sedentary rats. Nevertheless, EXE-EVs showed remarkable pro-angiogenic effect by augmenting proliferation, migration, and tube formation of HUVECs. Furthermore, the findings of animal experiments revealed that the EXE-EVs delivered by decellularized dermal matrix hydrogel (DDMH) could significantly promote the repair of skin defects through stimulating the regeneration of vascularized skin.Discussion: The present study is the first attempt to demonstrate that aerobic exercise-induced circulating EVs could be utilized as a cell-free therapy to activate angiogenesis and promote diabetic wound healing. Our findings suggest that EXE-EVs may stand for a potential strategy for diabetic soft tissue wound repair.
Collapse
Affiliation(s)
- Haifeng Liu
- Guangzhou Medical University, Guangzhou, China
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Bing Wu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xin Shi
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Yanpeng Cao
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Xin Zhao
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Daqiang Liang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Qihuang Qin
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Xinzhi Liang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Wei Lu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Daping Wang
- Guangzhou Medical University, Guangzhou, China
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- *Correspondence: Daping Wang, ; Jun Liu,
| | - Jun Liu
- Department of Limbs (Foot and Hand) Microsurgery, Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Chenzhou, China
- *Correspondence: Daping Wang, ; Jun Liu,
| |
Collapse
|
24
|
Lee-Ødegård S, Ueland T, Thorsby PM, Aukrust P, Michelsen AE, Halvorsen B, Drevon CA, Birkeland KI. Fetuin-A mediates the difference in adipose tissue insulin resistance between young adult pakistani and norwegian patients with type 2 diabetes. BMC Endocr Disord 2022; 22:208. [PMID: 35978354 PMCID: PMC9386965 DOI: 10.1186/s12902-022-01127-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND South-Asian immigrants to Western countries have a high prevalence of type 2 diabetes mellitus (T2DM) and increased adipose tissue insulin resistance (AT-IR), as compared to their Western counterparts. Fetuin-A is a hepatokine known to influence AT-IR. AIM Can plasma fetuin-A concentrations explain an ethnic difference in adipose tissue insulin resistance? METHODS We performed a two-step euglycemic-hyperinsulinaemic clamp and measured plasma concentrations of fetuin-A and non-esterified fatty acids (NEFA), in 18 Pakistani and 21 Norwegians with T2DM (age 29-45y) in Norway. AT-IR was calculated as NEFA-suppression during the clamp. The adipokines/cytokines leptin, adiponectin, visfatin, PTX3, IL-1β, INF-γ, and IL-4 were measured in fasting plasma. Liver fat was estimated by CT-scans. RESULTS Despite a lower BMI, Pakistani patients displayed higher AT-IR than Norwegians. NEFA-suppression during clamp was lower in Pakistani than Norwegians (mean=-20.6%, 95%CI=[-40.8, -0.01] and p = 0.046). Plasma fetuin-A concentration was higher in Pakistani than Norwegians (43.4 ng/mL[12.7,74.0], p = 0.007) and correlated negatively to %NEFA-suppression during clamp (rho=-0.39, p = 0.039). Plasma fetuin-A concentration explained 22% of the ethnic difference in NEFA-suppression during the clamp. Pakistani patients exhibited higher plasma leptin and lower PTX3 levels than Norwegian, and plasma visfatin correlated positively to plasma fetuin-A levels in the Pakistani patients. We observed no correlation between plasma fetuin-A and liver fat, but fetuin-A correlated negatively with plasma IL-1β, INF-γ, and IL-4 concentrations. Plasma IL-4 concentration was lower in Pakistani than in Norwegian patients. CONCLUSION Fetuin-A may contribute to explain the discrepancy in T2DM prevalence between Pakistani and Norwegians patients by influencing AT-IR.
Collapse
Affiliation(s)
| | - Thor Ueland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Per M Thorsby
- Hormone Laboratory, Dep of Medical Biochemistry and Biochemical endocrinology and metabolism research group, Oslo University Hospital, Aker, Oslo, Norway
| | - Pål Aukrust
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Annika E Michelsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kåre I Birkeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
25
|
Kim BJ. Effects of Muscles on Bone Metabolism—with a Focus on Myokines. Ann Geriatr Med Res 2022; 26:63-71. [PMID: 35722780 PMCID: PMC9271391 DOI: 10.4235/agmr.22.0054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/07/2022] [Indexed: 11/10/2022] Open
Abstract
Skeletal muscles and bones, the largest tissues in the body of a non-obese person, comprise the musculoskeletal system, which allows mobility and protects internal organs. Although muscles and bones are closely related throughout life, observations during development and aging and in human and animal disuse models have revealed the synchronization of tissue mass such that muscle phenotype changes precede alterations in bone mineral density and strength. This review discussed that mechanical forces, which have been the traditional research focus, are not the only mechanism by which muscle-derived signals may affect bone metabolism and emphasized the significance of skeletal muscles as an endocrine organ that secretes bone-regulatory factors. Consequently, both mechanical and biochemical aspects should be considered to fully understand muscle–bone crosstalk. This review also suggested that specific myokines could be ideal therapeutic targets for osteoporosis to both increase bone formation and reduce bone resorption; moreover, these myokines could also be potential circulating biomarkers to predict musculoskeletal health.
Collapse
Affiliation(s)
- Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
- Corresponding Authors: Beom-Jun Kim, MD, PhD Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea E-mail:
| |
Collapse
|
26
|
Jee H, Park E, Hur K, Kang M, Kim Y. High-Intensity Aerobic Exercise Suppresses Cancer Growth by Regulating Skeletal Muscle-Derived Oncogenes and Tumor Suppressors. Front Mol Biosci 2022; 9:818470. [PMID: 35801156 PMCID: PMC9254717 DOI: 10.3389/fmolb.2022.818470] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
High-intensity aerobic exercise (90% of the maximal heart rate) can effectively suppress cancer cell proliferation in vivo. However, the molecular effects of exercise and its relevance to cancer prevention remain uninvestigated. In this study, mice with colorectal cancer were subjected to high-intensity aerobic exercise, and mRNA-seq analysis was performed on the heart, lungs, and skeletal muscle tissues to analyze the genome-wide molecular effects of exercise. The skeletal muscle-derived genes with exercise-dependent differential expression were further evaluated for their effects on colorectal cancer cell viability. Compared to the results obtained for the control groups (healthy and cancer with no exercise), the regular and high-intensity aerobic physical activity in the mice produced positive results in comprehensive parameters (i.e., food intake, weight gain, and survival rate). A heatmap of differentially expressed genes revealed markedly different gene expression patterns among the groups. RNA-seq analysis of 23,282 genes expressed in the skeletal muscle yielded several anticancer effector genes (e.g., Trim63, Fos, Col1a1, and Six2). Knockdown and overexpression of selected anticancer genes repressed CT26 murine colorectal carcinoma cell proliferation by 20% (p < 0.05). Our findings, based on the aerobic exercise cancer mouse model, suggest that high-intensity aerobic exercise results in a comprehensive change in the expression patterns of genes, particularly those that can affect cancer cell viability. Such an approach may identify key exercise-regulated genes that can help the body combat cancer.
Collapse
Affiliation(s)
- Hyunseok Jee
- School of Kinesiology, Yeungnam University, Gyeongsan, South Korea
- College of Life Science and Nano Technology, Hannam University, Daejeon, South Korea
- *Correspondence: Hyunseok Jee, ; Yoosik Kim,
| | - Eunmi Park
- College of Life Science and Nano Technology, Hannam University, Daejeon, South Korea
| | - Kyunghoon Hur
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Minjeong Kang
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Yoosik Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Institute for Health Science and Technology (KIHST), KAIST, Daejeon, South Korea
- *Correspondence: Hyunseok Jee, ; Yoosik Kim,
| |
Collapse
|
27
|
Kirk EA, Castellani CA, Doherty TJ, Rice CL, Singh SM. Local and systemic transcriptomic responses from acute exercise induced muscle damage of the human knee extensors. Physiol Genomics 2022; 54:305-315. [PMID: 35723223 DOI: 10.1152/physiolgenomics.00146.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Skeletal muscle is adaptable to a direct stimulus of exercise-induced muscle damage (EIMD). Local muscle gene networks and systemic circulatory factors respond to EIMD within days, mediating anti-inflammation and cellular proliferation. Here we show in humans that local EIMD of one muscle group is associated with a systemic response of gene networks that regulate muscle structure and cellular development in non-local homologous muscle not directly altered by EIMD. In the non-dominant knee-extensors of seven males, EIMD was induced through voluntary contractions against an electric motor that lengthened muscles. Neuromuscular assessments, vastus lateralis muscle biopsies and blood draws occurred at two days prior, and one and two days post the EIMD intervention. From the muscle and blood plasma samples, RNA-seq measured transcriptome changes of differential expression using bioinformatic analyses.Relative to the time of the EIMD intervention, local muscle that was mechanically damaged had 475 genes differentially expressed, as compared to 33 genes in the non-local homologous muscle. Gene and network analysis showed that activity of the local muscle was related to structural maintenance, repair, and energetic processes, whereas gene and network activity of the non-local muscle (that was not directly modified by the EIMD) were related to muscle cell development, stress response, and structural maintenance. Altered expression of two novel miRNAs related to the EIMD response supported that systemic factors were active. Together, these results indicate that the expression of genes and gene networks that control muscle contractile structure can be modified in response to non-local EIMD in humans.
Collapse
Affiliation(s)
- Eric A Kirk
- School of Kinesiology, Faculty of Health Sciences, Western University, London, Ontario, Canada.,Molecular Genetics Unit, Department of Biology, Western University, London, Ontario, Canada
| | - Christina A Castellani
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Timothy J Doherty
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.,Department of Physical Medicine and Rehabilitation, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Charles L Rice
- School of Kinesiology, Faculty of Health Sciences, Western University, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Shiva M Singh
- Molecular Genetics Unit, Department of Biology, Western University, London, Ontario, Canada
| |
Collapse
|
28
|
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: 16.5] [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
|
29
|
Araújo MC, Soczek SHS, Pontes JP, Marques LAC, Santos GS, Simão G, Bueno LR, Maria-Ferreira D, Muscará MN, Fernandes ES. An Overview of the TRP-Oxidative Stress Axis in Metabolic Syndrome: Insights for Novel Therapeutic Approaches. Cells 2022; 11:cells11081292. [PMID: 35455971 PMCID: PMC9030853 DOI: 10.3390/cells11081292] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/19/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome (MS) is a complex pathology characterized by visceral adiposity, insulin resistance, arterial hypertension, and dyslipidaemia. It has become a global epidemic associated with increased consumption of high-calorie, low-fibre food and sedentary habits. Some of its underlying mechanisms have been identified, with hypoadiponectinemia, inflammation and oxidative stress as important factors for MS establishment and progression. Alterations in adipokine levels may favour glucotoxicity and lipotoxicity which, in turn, contribute to inflammation and cellular stress responses within the adipose, pancreatic and liver tissues, in addition to hepatic steatosis. The multiple mechanisms of MS make its clinical management difficult, involving both non-pharmacological and pharmacological interventions. Transient receptor potential (TRP) channels are non-selective calcium channels involved in a plethora of physiological events, including energy balance, inflammation and oxidative stress. Evidence from animal models of disease has contributed to identify their specific contributions to MS and may help to tailor clinical trials for the disease. In this context, the oxidative stress sensors TRPV1, TRPA1 and TRPC5, play major roles in regulating inflammatory responses, thermogenesis and energy expenditure. Here, the interplay between these TRP channels and oxidative stress in MS is discussed in the light of novel therapies to treat this syndrome.
Collapse
Affiliation(s)
- Mizael C. Araújo
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Suzany H. S. Soczek
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Jaqueline P. Pontes
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Maranhão, São Luís 565085-080, MA, Brazil;
| | - Leonardo A. C. Marques
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Gabriela S. Santos
- Programa de Pós-Graduação, Universidade CEUMA, São Luís 65075-120, MA, Brazil; (M.C.A.); (G.S.S.)
| | - Gisele Simão
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Laryssa R. Bueno
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
| | - Marcelo N. Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil; (L.A.C.M.); (M.N.M.)
| | - Elizabeth S. Fernandes
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil; (S.H.S.S.); (G.S.); (L.R.B.); (D.M.-F.)
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil
- Correspondence:
| |
Collapse
|
30
|
Lee-Ødegård S, Olsen T, Norheim F, Drevon CA, Birkeland KI. Potential Mechanisms for How Long-Term Physical Activity May Reduce Insulin Resistance. Metabolites 2022; 12:metabo12030208. [PMID: 35323652 PMCID: PMC8950317 DOI: 10.3390/metabo12030208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 02/20/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Insulin became available for the treatment of patients with diabetes 100 years ago, and soon thereafter it became evident that the biological response to its actions differed markedly between individuals. This prompted extensive research into insulin action and resistance (IR), resulting in the universally agreed fact that IR is a core finding in patients with type 2 diabetes mellitus (T2DM). T2DM is the most prevalent form of diabetes, reaching epidemic proportions worldwide. Physical activity (PA) has the potential of improving IR and is, therefore, a cornerstone in the prevention and treatment of T2DM. Whereas most research has focused on the acute effects of PA, less is known about the effects of long-term PA on IR. Here, we describe a model of potential mechanisms behind reduced IR after long-term PA to guide further mechanistic investigations and to tailor PA interventions in the therapy of T2DM. The development of such interventions requires knowledge of normal glucose metabolism, and we briefly summarize an integrated physiological perspective on IR. We then describe the effects of long-term PA on signaling molecules involved in cellular responses to insulin, tissue-specific functions, and whole-body IR.
Collapse
Affiliation(s)
- Sindre Lee-Ødegård
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
| | - Christian Andre Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway; (T.O.); (F.N.); (C.A.D.)
- Vitas Ltd. Analytical Services, Oslo Science Park, 0349 Oslo, Norway
| | - Kåre Inge Birkeland
- Department of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway;
- Correspondence:
| |
Collapse
|
31
|
Single bout of exercise triggers the increase of vitamin D blood concentration in adolescent trained boys: a pilot study. Sci Rep 2022; 12:1825. [PMID: 35115578 PMCID: PMC8814171 DOI: 10.1038/s41598-022-05783-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Vitamin D is necessary for musculoskeletal health, however, the supplementation of vitamin D above the sufficiency level does not bring additional bone mass density (BMD), unlike physical exercise which enhances the bone formatting process. Regular physical activity has been shown to upregulate VDR expression in muscles and to increase circulating vitamin D. Here we investigate whether a single bout of exercise might change 25(OH)D3 blood concentration and how it affects metabolic response to exercise. Twenty-six boys, 13.8 years old (SD ± 0.7) soccer players, participated in the study. The participants performed one of two types of exercise: the first group performed the VO2max test until exhaustion, and the second performed three times the repeated 30 s Wingate Anaerobic Test (WAnT). Blood was collected before, 15 min and one hour after the exercise. The concentration of 25(OH)D3, parathyroid hormone (PTH), interleukin-6 (IL-6), lactate, non-esterified fatty acids (NEFA) and glycerol were determined. 25(OH)D3 concentration significantly increased after the exercise in all boys. The most prominent changes in 25(OH)D3, observed after WAnT, were associated with the rise of PTH. The dimensions of response to the exercises observed through the changes in the concentration of 25(OH)D3, PTH, NEFA and glycerol were associated with the significant increases of IL-6 level. A single bout of exercise may increase the serum’s 25(OH)D3 concentration in young trained boys. The intensive interval exercise brings a more potent stimulus to vitamin D fluctuations in young organisms. Our results support the hypothesis that muscles may both store and release 25(OH)D3.
Collapse
|
32
|
Cortiula F, Hendriks LEL, van de Worp WRPH, Schols AMWJ, Vaes RDW, Langen RCJ, De Ruysscher D. Physical exercise at the crossroad between muscle wasting and the immune system: implications for lung cancer cachexia. J Cachexia Sarcopenia Muscle 2022; 13:55-67. [PMID: 35014216 PMCID: PMC8818640 DOI: 10.1002/jcsm.12900] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/11/2021] [Accepted: 11/28/2021] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a syndrome characterized by involuntary weight loss and wasting of skeletal muscle mass. It is associated with worse overall survival and quality of life. The cancer-induced systemic inflammation and the consequent host derived catabolic stimuli, trigger cachexia by inhibiting muscle protein synthesis and enhancing muscle catabolism. The muscle itself may further promote chronic inflammation, introducing a vicious catabolic circle. Nutritional support alone plays a limited role in the treatment of cancer cachexia and should be combined with other interventions. Physical exercise lowers systemic inflammation and promotes muscle anabolism. It also attenuates the age-related physical decline in elderly and it might counteract the muscle wasting induced by the cancer cachexia syndrome. This review describes how cancer-induced systemic inflammation promotes muscle wasting and whether physical exercise may represent a suitable treatment for cancer-induced cachexia, particularly in patients with non-small cell lung cancer. We summarized pre-clinical and clinical studies investigating whether physical exercise would improve muscle performance and whether this improvement would translate in a clinically meaningful benefit for patients with cancer, in terms of survival and quality of life. Moreover, this review describes the results of studies investigating the interplay between physical exercise and the immune system, including the role of the intestinal microbiota.
Collapse
Affiliation(s)
- Francesco Cortiula
- Department of Radiation Oncology (MAASTRO), Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Lizza E L Hendriks
- Department of Respiratory Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center +, Maastricht, The Netherlands
| | - Wouter R P H van de Worp
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Annemie M W J Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Rianne D W Vaes
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ramon C J Langen
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
33
|
Abstract
Physical exercise can be effective in preventing or ameliorating various diseases, including diabetes, cardiovascular diseases, neurodegenerative diseases, and cancer. However, not everyone may be able to participate in exercise due to illnesses, age-related frailty, or difficulty in long-term behavior change. An alternative option is to utilize pharmacological interventions that mimic the positive effects of exercise training. Recent studies have identified signaling pathways associated with the benefits of physical activity and discovered exercise mimetics that can partially simulate the systemic impact of exercise. This review describes the molecular targets for exercise mimetics and their effect on skeletal muscle and other tissues. We will also discuss the potential advantages of using natural products as a multi-targeting agent for mimicking the health-promoting effects of exercise.
Collapse
Affiliation(s)
- Young Jin Jang
- Major of Food Science & Technology, Seoul Women’s University, Seoul 01797, Korea
| | - Sanguine Byun
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
34
|
Al-Rawaf HA, Alghadir AH, Gabr SA. Expression of Circulating MicroRNAs and Myokines and Interactions with Serum Osteopontin in Type 2 Diabetic Patients with Moderate and Poor Glycemic Control: A Biochemical and Molecular Study. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7453000. [PMID: 34917685 PMCID: PMC8670937 DOI: 10.1155/2021/7453000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 11/15/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Cellular miRNAs are expressed in tissue fluids with sufficient amounts and were identified as potential molecular targets for studying the physiological mechanisms and correlations with many human diseases particularly diabetes. However, molecular-based changes among older adults with diabetes mellitus (DM) are rarely fully elucidated. AIM This study is aimed at identifying circulating miRNAs, which hold the potential to serve as biomarkers for the immune-inflammatory changes in older T2D patients with moderate and poor glycemic control status. In addition, the association of both myokines and osteopontin (OPN) levels with circulating miRNAs was identified. METHODS A total of 80 subjects aged 20-80 years were invited during the period of October 2017-May 2018 to participate in this descriptive cross-sectional study. All subjects were diagnosed with T2D for more than 5 years. Subjects were grouped based on glycemic control (HbA1c values) into two groups: moderate glycemic control (>7-8% HbA1c, no = 30) and poor glycemic control (>8% HbA1c, no = 50), respectively. Diabetic control parameters, fasting blood sugar (FS), HbA1c, fasting insulin (IF), insulin resistance (IR), HOMA-IR, inflammatory cytokines (IL-6, IL-8, IL-18, IL-23, TNF-α, and CRP), osteopontin, and myokines (adropin and irisin) were estimated by colorimetric and immune ELISA assays, respectively. In addition, real-time RT-PCR analysis was performed to evaluate the expression of circulating miRNAs, miR-146a and miR-144, in the serum of all diabetic subjects. RESULTS In this study, T2D patients with poor glycemic control showed a significant increase in the serum levels of IL-6, IL-8, IL-18, IL-23, TNF-α, CRP, and OPN and a reduction in the levels of myokines, adropin and irisin, compared to patients with moderate glycemic control. The results obtained are significantly correlated with the severity of diabetes measured by HbA1c, FS, IF, and HOMA-IR. In addition, baseline expression of miR-146a is significantly reduced and miR-144 is significantly increased in T2D patients with poor glycemic control compared to those with moderate glycemic control. In all diabetic groups, the expression of miR-146a and miR-144 is significantly correlated with diabetic controls, inflammatory cytokines, myokines, and serum levels of OPN. Respective of gender, women with T2D showed more significant change in the expressed miRNAs, inflammatory cytokines, OPN, and serum myokine markers compared to men. ROC analysis identified AUC cutoff values of miR-146a, miR-144, adropin, irisin, and OPN expression levels with considerable specificity and sensitivity which recommends the potential use of adropin, irisin, and OPN as diagnostic biomarkers for diabetes with varying glycemic control status. CONCLUSION In this study, molecular expression of certain microRNA species, such as miR-146a and miR-144, was identified and significantly associated with parameters of disease severity, HbA1c, inflammatory cytokines, myokines, and serum osteopontin in T2D patients with moderate and poor glycemic control. The AUC cutoff values of circulating miRNAs, miR-146a and miR-144; myokines, adropin and irisin; and serum OPN were significantly identified by ROC analysis which additionally recommends the potential use of these biomarkers, miR-146a, miR-144, adropin, irisin, and OPN, as diagnostic biomarkers with considerable specificity and sensitivity for diabetes in patients with varying glycemic control status.
Collapse
Affiliation(s)
- Hadeel A. Al-Rawaf
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Ahmad H. Alghadir
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Sami A. Gabr
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| |
Collapse
|
35
|
Secretome from In Vitro Mechanically Loaded Myoblasts Induces Tenocyte Migration, Transition to a Fibroblastic Phenotype and Suppression of Collagen Production. Int J Mol Sci 2021; 22:ijms222313089. [PMID: 34884895 PMCID: PMC8657858 DOI: 10.3390/ijms222313089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
It is known that mechanical loading of muscles increases the strength of healing tendon tissue, but the mechanism involved remains elusive. We hypothesized that the secretome from myoblasts in co-culture with tenocytes affects tenocyte migration, cell phenotype, and collagen (Col) production and that the effect is dependent on different types of mechanical loading of myoblasts. To test this, we used an in vitro indirect transwell co-culture system. Myoblasts were mechanically loaded using the FlexCell® Tension system. Tenocyte cell migration, proliferation, apoptosis, collagen production, and several tenocyte markers were measured. The secretome from myoblasts decreased the Col I/III ratio and increased the expression of tenocyte specific markers as compared with tenocytes cultured alone. The secretome from statically loaded myoblasts significantly enhanced tenocyte migration and Col I/III ratio as compared with dynamic loading and controls. In addition, the secretome from statically loaded myoblasts induced tenocytes towards a myofibroblast-like phenotype. Taken together, these results demonstrate that the secretome from statically loaded myoblasts has a profound influence on tenocytes, affecting parameters that are related to the tendon healing process.
Collapse
|
36
|
Williams K, Carrasquilla GD, Ingerslev LR, Hochreuter MY, Hansson S, Pillon NJ, Donkin I, Versteyhe S, Zierath JR, Kilpeläinen TO, Barrès R. Epigenetic rewiring of skeletal muscle enhancers after exercise training supports a role in whole-body function and human health. Mol Metab 2021; 53:101290. [PMID: 34252634 PMCID: PMC8355925 DOI: 10.1016/j.molmet.2021.101290] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives Regular physical exercise improves health by reducing the risk of a plethora of chronic disorders. We hypothesized that endurance exercise training remodels the activity of gene enhancers in skeletal muscle and that this remodeling contributes to the beneficial effects of exercise on human health. Methods and results By studying changes in histone modifications, we mapped the genome-wide positions and activities of enhancers in skeletal muscle biopsies collected from young sedentary men before and after 6 weeks of endurance exercise. We identified extensive remodeling of enhancer activities after exercise training, with a large subset of the remodeled enhancers located in the proximity of genes transcriptionally regulated after exercise. By overlapping the position of enhancers with genetic variants, we identified an enrichment of disease-associated genetic variants within the exercise-remodeled enhancers. Conclusion Our data provide evidence of a functional link between epigenetic rewiring of enhancers to control their activity after exercise training and the modulation of disease risk in humans. Exercise training changes in skeletal muscle gene expression is enriched for secreted factors. The activity of skeletal muscle enhancers undergoes substantial remodeling after exercise training. Skeletal muscle enhancer activity and gene transcription are strongly associated. Exercise training-remodeled enhancer regions are enriched for GWAS SNPs associated with human traits and diseases.
Collapse
Affiliation(s)
- Kristine Williams
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Germán D Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Lars Roed Ingerslev
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Mette Yde Hochreuter
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Svenja Hansson
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Nicolas J Pillon
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Ida Donkin
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Soetkin Versteyhe
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark; Department of Physiology and Pharmacology, Karolinska Institutet, 171 77, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark.
| |
Collapse
|
37
|
Vepkhvadze TF, Vorotnikov AV, Popov DV. Electrical Stimulation of Cultured Myotubes in vitro as a Model of Skeletal Muscle Activity: Current State and Future Prospects. BIOCHEMISTRY (MOSCOW) 2021; 86:597-610. [PMID: 33993862 DOI: 10.1134/s0006297921050084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Skeletal muscles comprise more than a third of human body mass and critically contribute to regulation of body metabolism. Chronic inactivity reduces metabolic activity and functional capacity of muscles, leading to metabolic and other disorders, reduced life quality and duration. Cellular models based on progenitor cells isolated from human muscle biopsies and then differentiated into mature fibers in vitro can be used to solve a wide range of experimental tasks. The review discusses the aspects of myogenesis dynamics and regulation, which might be important in the development of an adequate cell model. The main function of skeletal muscle is contraction; therefore, electrical stimulation is important for both successful completion of myogenesis and in vitro modeling of major processes induced in the skeletal muscle by acute or regular physical exercise. The review analyzes the drawbacks of such cellular model and possibilities for its optimization, as well as the prospects for its further application to address fundamental aspects of muscle physiology and biochemistry and explore cellular and molecular mechanisms of metabolic diseases.
Collapse
Affiliation(s)
- Tatiana F Vepkhvadze
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia
| | - Alexander V Vorotnikov
- National Medical Research Center of Cardiology, Ministry of Healthcare of the Russian Federation, Moscow, 121552, Russia
| | - Daniil V Popov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia. .,Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
38
|
Droujinine IA, Meyer AS, Wang D, Udeshi ND, Hu Y, Rocco D, McMahon JA, Yang R, Guo J, Mu L, Carey DK, Svinkina T, Zeng R, Branon T, Tabatabai A, Bosch JA, Asara JM, Ting AY, Carr SA, McMahon AP, Perrimon N. Proteomics of protein trafficking by in vivo tissue-specific labeling. Nat Commun 2021; 12:2382. [PMID: 33888706 PMCID: PMC8062696 DOI: 10.1038/s41467-021-22599-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Conventional approaches to identify secreted factors that regulate homeostasis are limited in their abilities to identify the tissues/cells of origin and destination. We established a platform to identify secreted protein trafficking between organs using an engineered biotin ligase (BirA*G3) that biotinylates, promiscuously, proteins in a subcellular compartment of one tissue. Subsequently, biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Applying this approach in Drosophila, we identify 51 muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, including CG2145 (human ortholog ENDOU) that binds directly to muscles and promotes activity. In addition, in mice, we identify 291 serum proteins secreted from conditional BirA*G3 embryo stem cell-derived teratomas, including low-abundance proteins with hormonal properties. Our findings indicate that the communication network of secreted proteins is vast. This approach has broad potential across different model systems to identify cell-specific secretomes and mediators of interorgan communication in health or disease.
Collapse
Affiliation(s)
- Ilia A Droujinine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, USA.
| | - Amanda S Meyer
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Dan Wang
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Entomology, China Agricultural University, Beijing, China
| | | | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - David Rocco
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Rui Yang
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - JinJin Guo
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Luye Mu
- Department of Electrical Engineering, Yale University, New Haven, CT, USA
| | | | | | - Rebecca Zeng
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Tess Branon
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Areya Tabatabai
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Justin A Bosch
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - John M Asara
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Alice Y Ting
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Departments of Genetics, Biology, and Chemistry, Stanford University, Stanford, CA, USA
| | - Steven A Carr
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
39
|
Methods for Proteomics-Based Analysis of the Human Muscle Secretome Using an In Vitro Exercise Model. Methods Mol Biol 2021; 2261:433-442. [PMID: 33421006 DOI: 10.1007/978-1-0716-1186-9_27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Over the last decade, the skeletal muscle as a secretory organ has gained importance. A growing number of peptides is described which are produced and released by the muscle fibers and work in an autocrine, paracrine, and endocrine fashion. The contraction-induced secretion of these myokines is considered to contribute to the health promoting effects of exercise. To gain further insights into the molecular processes that occur during contraction, an in vitro exercise model, electric pulse stimulation (EPS), was established. Recent publications show that this model is suitable to electrostimulate human skeletal muscle cells and thus mimic muscle contraction in vitro. Here, we provide a detailed protocol for the proteomics-based analysis of the human muscle secretome, starting with the cultivation of human myotubes and ending with sample preparation for targeted and untargeted proteome analysis of the cell culture supernatant. This workflow should allow for deeper insights into the complex nature of the muscle secretome and the identification of new myokines which might help to understand the cross talk of the working muscle with different organs and the beneficial effects of exercise.
Collapse
|
40
|
Onat T, Inandiklioglu N. Circulating Myonectin and Irisin Levels in Gestational Diabetes Mellitus - A Case-control Study. Z Geburtshilfe Neonatol 2021; 225:320-326. [PMID: 33728620 DOI: 10.1055/a-1404-4323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The objective of this study was to assess the levels of serum myonectin and irisin in pregnant women with and without gestational diabetes mellitus (GDM). METHOD A total of 80 pregnant women participated in our study (which consisted of 40 patients with GDM, 40 participants as the control group). Myonectin and irisin levels were analyzed through the ELISA technique, in addition to metabolic parameters in the serum samples of the participants. RESULTS It was found that the levels of irisin and myonectin were lower in the GDM group compared to the control group. Moreover, it was determined that the values of age (p<0.001), body mass index (p=0.001), gravida (p=0.001), parity (p = 0.016), fasting serum glucose (p=0.001), fasting serum insulin (p=0.007), postprandial serum glucose (p=0.006), HbA1c (p<0.001), HOMA-IR (p<0.001) were higher; HDL cholesterol (p<0.001) was lower. Insulin resistance was significantly higher in the GDM group. CONCLUSIONS Levels of myonectin and irisin were determined to be low in the GDM group. Our results have demonstrated that myonectin and irisin could play a role in the development of GDM and that irisin as well as myonectin could be a novel biomarker for GDM.
Collapse
Affiliation(s)
- Taylan Onat
- Department of Obstetrics and Gynecology, Bozok University Faculty of Medicine, Yozgat, Turkey
| | | |
Collapse
|
41
|
Nederveen JP, Warnier G, Di Carlo A, Nilsson MI, Tarnopolsky MA. Extracellular Vesicles and Exosomes: Insights From Exercise Science. Front Physiol 2021; 11:604274. [PMID: 33597890 PMCID: PMC7882633 DOI: 10.3389/fphys.2020.604274] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/10/2020] [Indexed: 12/20/2022] Open
Abstract
The benefits of exercise on health and longevity are well-established, and evidence suggests that these effects are partially driven by a spectrum of bioactive molecules released into circulation during exercise (e.g., exercise factors or 'exerkines'). Recently, extracellular vesicles (EVs), including microvesicles (MVs) and exosomes or exosome-like vesicles (ELVs), were shown to be secreted concomitantly with exerkines. These EVs have therefore been proposed to act as cargo carriers or 'mediators' of intercellular communication. Given these findings, there has been a rapidly growing interest in the role of EVs in the multi-systemic, adaptive response to exercise. This review aims to summarize our current understanding of the effects of exercise on MVs and ELVs, examine their role in the exercise response and long-term adaptations, and highlight the main methodological hurdles related to blood collection, purification, and characterization of ELVs.
Collapse
Affiliation(s)
- Joshua P Nederveen
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Geoffrey Warnier
- Institut of Neuroscience, UCLouvain, Université catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Alessia Di Carlo
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Mats I Nilsson
- Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| | - Mark A Tarnopolsky
- Department of Pediatrics, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada.,Exerkine Corporation, McMaster University Medical Centre (MUMC), Hamilton, ON, Canada
| |
Collapse
|
42
|
Mahyudin F, Yazid H, Edward M, Basuki MH, Bari YA, Rantam FA. The enhancement apoptosis of osteosarcoma mesenchymal stem cells co-cultivation with peripheral blood mononuclear cells sensitized by secretome and granulocyte macrophage colony-stimulating factor. J Adv Pharm Technol Res 2020; 11:213-219. [PMID: 33425707 PMCID: PMC7784941 DOI: 10.4103/japtr.japtr_52_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/06/2020] [Accepted: 08/25/2020] [Indexed: 12/22/2022] Open
Abstract
The advanced, metastasis, and reccurent of osteosarcoma (OS) patients have a poor prognosis postaggresive surgery and chemotherapy. Peripheral blood mononuclear cells (PBMCs) as cell-based immunotherapy may successful in the OS treatment. To investigate the enhancement apoptosis of OS-mesenchymal stem cells (OS-MSCs) co-cultivated with PBMCs sensitized using the secretome and granulocyte macrophage colony-stimulating factor (GMCSF). This true experimental study with posttest only control group design and in vitro study. The sample was cultured OS-MSCs which confirmed by Cluster of Differentiation-133 using immunocytochemistry (ICC) and histopathology analysis. The sample divided into six groups accordingly: OS-MSC, OS-MSC + PMBC, OS-MSC + PMBC + Secretome, OS-MSC + PMBC + GMCSF, OS-MSC + PBMC + Secretome + GMCSF (n = 5/N = 30). The enhancement of OS-MSCs apoptosis was analyzed through Interleukin-2 (IL-2) level through the Enyzme-Linked Immunosorbent Assay examination, expression of Signal Transducers and Activators of Transcription (STAT)-3 and caspase-3 by ICC. One-way analysis of variance test and Tukey Honestly Significant Difference to analyze the difference between the groups (P < 0.05). The highest of IL-2 level was found in the PBMC + Secretome + GMCSF group. The highest expression of caspase-3 was found in OS-MSC + PBMC + Secretome + GMCSF group with significant different between groups (P < 0.05). There was insignificant difference of STAT-3 epxression and IL-2 level between groups (P > 0.05). The co-cultivation of OS-MSCs and PBMSCs activated using secretome and GMCSF has a great ability to enhance OS-MSCs apoptosis.
Collapse
Affiliation(s)
- Ferdiansyah Mahyudin
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Hizbillah Yazid
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Mouli Edward
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Mohammad Hardian Basuki
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Yunus Abdul Bari
- Orthopedic and Traumatology Department, Faculty of Medicine, Dr Soetomo General Hospital, Airlangga University, Surabaya, Indonesia
| | - Fedik Abdul Rantam
- Stem Cell Research And Development Center, Airlangga University, Surabaya, Indonesia
| |
Collapse
|
43
|
Harten IA, Kaber G, Agarwal KJ, Kang I, Ibarrientos SR, Workman G, Chan CK, Nivison MP, Nagy N, Braun KR, Kinsella MG, Merrilees MJ, Wight TN. The synthesis and secretion of versican isoform V3 by mammalian cells: A role for N-linked glycosylation. Matrix Biol 2020; 89:27-42. [PMID: 32001344 PMCID: PMC7282976 DOI: 10.1016/j.matbio.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 02/02/2023]
Abstract
Versican is a large extracellular matrix (ECM) chondroitin sulfate (CS) proteoglycan found in most soft tissues, which is encoded by the VCAN gene. At least four major isoforms (V0, V1, V2, and V3) are generated via alternative splicing. The isoforms of versican are expressed and accumulate in various tissues during development and disease, where they contribute to ECM structure, cell growth and migration, and immune regulation, among their many functions. While several studies have identified the mRNA transcript for the V3 isoform in a number of tissues, little is known about the synthesis, secretion, and targeting of the V3 protein. In this study, we used lentiviral generation of doxycycline-inducible rat V3 with a C-terminal tag in stable NIH 3T3 cell lines and demonstrated that V3 is processed through the classical secretory pathway. We further show that N-linked glycosylation is required for efficient secretion and solubility of the protein. By site-directed mutagenesis, we identified amino acids 57 and 330 as the active N-linked glycosylation sites on V3 when expressed in this cell type. Furthermore, exon deletion constructs of V3 revealed that exons 11-13, which code for portions of the carboxy region of the protein (G3 domain), are essential for V3 processing and secretion. Once secreted, the V3 protein associates with hyaluronan along the cell surface and within the surrounding ECM. These results establish critical parameters for the processing, solubility, and targeting of the V3 isoform by mammalian cells and establishes a role for V3 in the organization of hyaluronan.
Collapse
Affiliation(s)
- Ingrid A. Harten
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Gernot Kaber
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Kiran J. Agarwal
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Inkyung Kang
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | | | - Gail Workman
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Christina K. Chan
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Mary P. Nivison
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Nadine Nagy
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Kathleen R. Braun
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | | | - Mervyn J. Merrilees
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Thomas N. Wight
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| |
Collapse
|
44
|
Gut microbiota and regulation of myokine-adipokine function. Curr Opin Pharmacol 2020; 52:9-17. [DOI: 10.1016/j.coph.2020.03.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/26/2020] [Accepted: 03/28/2020] [Indexed: 12/27/2022]
|
45
|
Florin A, Lambert C, Sanchez C, Zappia J, Durieux N, Tieppo AM, Mobasheri A, Henrotin Y. The secretome of skeletal muscle cells: A systematic review. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 2:100019. [DOI: 10.1016/j.ocarto.2019.100019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 12/18/2019] [Indexed: 12/18/2022] Open
|
46
|
Kim G, Kim JH. Impact of Skeletal Muscle Mass on Metabolic Health. Endocrinol Metab (Seoul) 2020; 35:1-6. [PMID: 32207258 PMCID: PMC7090295 DOI: 10.3803/enm.2020.35.1.1] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle is regarded as an endocrine and paracrine organ. Muscle-derived secretory proteins, referred to as myokines, mediate interactions between skeletal muscle mass and other organs such as the liver, adipose tissue, pancreas, bone, and the cardiovascular system. As individuals age, reduced levels of physical activity and sarcopenia (loss of skeletal muscle mass and strength) are associated with physical frailty and disability. Recently, several studies have suggested that the loss of skeletal muscle mass may contribute to metabolic disease. Therefore, herein, we focus on the relationships between skeletal muscle mass and metabolic diseases, including metabolic syndrome and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Gyuri Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Hyeon Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
47
|
Vechetti IJ, Valentino T, Mobley CB, McCarthy JJ. The role of extracellular vesicles in skeletal muscle and systematic adaptation to exercise. J Physiol 2020; 599:845-861. [PMID: 31944292 DOI: 10.1113/jp278929] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022] Open
Abstract
Regular exercise has a central role in human health by reducing the risk of type 2 diabetes, obesity, stroke and cancer. How exercise is able to promote such systemic benefits has remained somewhat of a mystery but has been thought to be in part mediated by the release of myokines, skeletal muscle-specific cytokines, in response to exercise. Recent studies have revealed skeletal muscle can also release extracellular vesicles (EVs) into circulation following a bout of exercise. EVs are small membrane-bound vesicles capable of delivering biomolecules to recipient cells and subsequently altering their metabolism. The notion that EVs may have a role in both skeletal muscle and systemic adaptation to exercise has generated a great deal of excitement within a number of different fields including exercise physiology, neuroscience and metabolism. The purpose of this review is to provide an introduction to EV biology and what is currently known about skeletal muscle EVs and their potential role in the response of muscle and other tissues to exercise.
Collapse
Affiliation(s)
- Ivan J Vechetti
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Taylor Valentino
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - C Brooks Mobley
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - John J McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, 40536, USA
| |
Collapse
|
48
|
Laurens C, Bergouignan A, Moro C. Exercise-Released Myokines in the Control of Energy Metabolism. Front Physiol 2020; 11:91. [PMID: 32116795 PMCID: PMC7031345 DOI: 10.3389/fphys.2020.00091] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 01/27/2020] [Indexed: 12/22/2022] Open
Abstract
Physical activity reduces cardiometabolic risk, while physical inactivity increases chronic diseases risk. This led to the idea that exercise-induced muscle contraction contributes to metabolic regulation and health. It is now well established that skeletal muscle, through the release of endocrine factors, i.e., so-called myokines, crosstalk with metabolic organs such as adipose tissue, liver and pancreas. Recent advances suggested that a number of myokines are able to modulate adipose tissue metabolism and thermogenic activity, liver endogenous glucose production and β-cell insulin secretion. This novel paradigm offers a compelling hypothesis and molecular basis to explain the link between physical inactivity and chronic diseases. Herein, we review major findings and recent advances linking exercise, myokines secretion and inter-organ crosstalk. Identifying the molecular mediators linking physical activity to metabolic health could open the path toward novel therapeutic targets in metabolic diseases.
Collapse
Affiliation(s)
- Claire Laurens
- CNRS, IPHC, UMR 7178, Université de Strasbourg, Strasbourg, France.,Centre National d'Etudes Spatiales, Paris, France
| | - Audrey Bergouignan
- CNRS, IPHC, UMR 7178, Université de Strasbourg, Strasbourg, France.,Division of Endocrinology, Metabolism and Diabetes, Anschutz Health & Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Cedric Moro
- INSERM, UMR 1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Paul Sabatier University, University of Toulouse, Toulouse, France
| |
Collapse
|
49
|
Targeting Age-Dependent Functional and Metabolic Decline of Human Skeletal Muscle: The Geroprotective Role of Exercise, Myokine IL-6, and Vitamin D. Int J Mol Sci 2020; 21:ijms21031010. [PMID: 32033000 PMCID: PMC7037081 DOI: 10.3390/ijms21031010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 02/06/2023] Open
Abstract
In the elderly, whole-body health largely relies on healthy skeletal muscle, which controls body stability, locomotion, and metabolic homeostasis. Age-related skeletal muscle structural/functional deterioration is associated with a higher risk of severe comorbid conditions and poorer outcomes, demanding major socioeconomic costs. Thus, the need for efficient so-called geroprotective strategies to improve resilience and ensure a good quality of life in older subjects is urgent. Skeletal muscle senescence and metabolic dysregulation share common cellular/intracellular mechanisms, potentially representing targets for intervention to preserve muscle integrity. Many factors converge in aging, and multifaceted approaches have been proposed as interventions, although they have often been inconclusive. Physical exercise can counteract aging and metabolic deficits, not only in maintaining tissue mass, but also by preserving tissue secretory function. Indeed, skeletal muscle is currently considered a proper secretory organ controlling distant organ functions through immunoactive regulatory small peptides called myokines. This review provides a current perspective on the main biomolecular mechanisms underlying age-dependent and metabolic deterioration of skeletal muscle, herein discussed as a secretory organ, the functional integrity of which largely depends on exercise and myokine release. In particular, muscle-derived interleukin (IL)-6 is discussed as a nutrient-level biosensor. Overall, exercise and vitamin D are addressed as optimal geroprotective strategies in view of their multi-target effects.
Collapse
|
50
|
Kitakaze T, Yoshikawa M, Kobayashi Y, Kimura N, Goshima N, Ishikawa T, Ogata Y, Yamashita Y, Ashida H, Harada N, Yamaji R. Extracellular transglutaminase 2 induces myotube hypertrophy through G protein-coupled receptor 56. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118563. [PMID: 31666191 DOI: 10.1016/j.bbamcr.2019.118563] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 12/15/2022]
Abstract
Skeletal muscle secretes biologically active proteins that contribute to muscle hypertrophy in response to either exercise or dietary intake. The identification of skeletal muscle-secreted proteins that induces hypertrophy can provide critical information regarding skeletal muscle health. Dietary provitamin A, β-carotene, induces hypertrophy of the soleus muscle in mice. Here, we hypothesized that skeletal muscle produces hypertrophy-inducible secretory proteins via dietary β-carotene. Knockdown of retinoic acid receptor (RAR) γ inhibited the β-carotene-induced increase soleus muscle mass in mice. Using RNA sequencing, bioinformatic analyses, and literature searching, we predicted transglutaminase 2 (TG2) to be an all-trans retinoic acid (ATRA)-induced secretory protein in cultured C2C12 myotubes. Tg2 mRNA expression increased in ATRA- or β-carotene-stimulated myotubes and in the soleus muscle of β-carotene-treated mice. Knockdown of RARγ inhibited β-carotene-increased mRNA expression of Tg2 in the soleus muscle. ATRA increased endogenous TG2 levels in conditioned medium from myotubes. Extracellular TG2 promoted the phosphorylation of Akt, mechanistic target of rapamycin (mTOR), and ribosomal p70 S6 kinase (p70S6K), and inhibitors of mTOR, phosphatidylinositol 3-kinase, and Src (rapamycin, LY294002, and Src I1, respectively) inhibited TG2-increased phosphorylation of mTOR and p70S6K. Furthermore, extracellular TG2 promoted protein synthesis and hypertrophy in myotubes. TG2 mutant lacking transglutaminase activity exerted the same effects as wild-type TG2. Knockdown of G protein-coupled receptor 56 (GPR56) inhibited the effects of TG2 on mTOR signaling, protein synthesis, and hypertrophy. These results indicated that TG2 expression was upregulated through ATRA-mediated RARγ and that extracellular TG2 induced myotube hypertrophy by activating mTOR signaling-mediated protein synthesis through GPR56, independent of transglutaminase activity.
Collapse
MESH Headings
- Animals
- Cell Enlargement/drug effects
- Cell Line
- GTP-Binding Proteins/genetics
- GTP-Binding Proteins/metabolism
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Mice
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Myoblasts/cytology
- Myoblasts/metabolism
- Phosphorylation/drug effects
- Protein Glutamine gamma Glutamyltransferase 2
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Retinoic Acid/antagonists & inhibitors
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Retinoic Acid Receptor alpha/antagonists & inhibitors
- Retinoic Acid Receptor alpha/genetics
- Retinoic Acid Receptor alpha/metabolism
- Ribosomal Protein S6 Kinases, 70-kDa/metabolism
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/metabolism
- Transglutaminases/genetics
- Transglutaminases/metabolism
- Tretinoin/pharmacology
- beta Carotene/administration & dosage
- beta Carotene/pharmacology
- Retinoic Acid Receptor gamma
Collapse
Affiliation(s)
- Tomoya Kitakaze
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan; Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | - Miki Yoshikawa
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Yasuyuki Kobayashi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Naohiro Kimura
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Naoki Goshima
- National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Takahiro Ishikawa
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, Matsue, Shimane, Japan
| | - Yoshiyuki Ogata
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Yoko Yamashita
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | - Hitoshi Ashida
- Department of Agrobioscience, Graduate School of Agricultural Science, Kobe University, Kobe, Hyogo, Japan
| | - Naoki Harada
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Ryoichi Yamaji
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan.
| |
Collapse
|