1
|
Jaiswal AK, Kushawaha AK, Pawan Kumar, Ansari A, Chhikara N, Hemlata Bhatt, Katiyar S, Ahmad I, Choudhury AD, Bhatta RS, Tamrakar AK, Sashidhara KV. Design, synthesis, and biological evaluation of quinazolinone-dihydropyrimidinone as a potential anti-diabetic agent via GLUT4 translocation stimulation. Eur J Med Chem 2025; 288:117366. [PMID: 39954347 DOI: 10.1016/j.ejmech.2025.117366] [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: 12/02/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/17/2025]
Abstract
A library of 30 novel quinazolinone-dihydropyrimidinone derivatives was synthesized employing a diversity-oriented approach for the identification of potential anti-diabetic therapeutic lead. In vitro evaluation revealed that seven compounds (5d, 5e, 5i, 5j, 5l, 5m and 5s) significantly enhanced the rate of GLUT4 translocation to the cell surface in L6-GLUT4myc myotubes. Out of these, compound, 5m exhibited promising potency to stimulate GLUT4 translocation in skeletal muscle cells via activating AMPK-dependent pathway, but independent to PI-3-K/AKT signaling. Under in vivo conditions, treatment with 5m demonstrated a marked 39.5 % (p < 0.001) reduction in blood glucose levels in a streptozotocin-induced diabetic rat model after 5 h of treatment. Pharmacokinetic analysis indicated compound 5m shows favourable pharmacokinetic properties. Overall, the compound 5m emerged as a promising lead compound for subsequent structural modification and optimization to develop a novel and potent anti-hyperglycemic agent.
Collapse
Affiliation(s)
- Arvind Kumar Jaiswal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Ajay Kishor Kushawaha
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Pawan Kumar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Alisha Ansari
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Nikita Chhikara
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Hemlata Bhatt
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Sarita Katiyar
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Ishbal Ahmad
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Abhijit Deb Choudhury
- Pharmaceutics and Pharmacokinetics Division, CSIR- Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Rabi Sankar Bhatta
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India; Pharmaceutics and Pharmacokinetics Division, CSIR- Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Akhilesh K Tamrakar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| | - Koneni V Sashidhara
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute BS, 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India; Sophisticated Analytical Instrument Facility & Research, CSIR-Central Drug Research Institute, BS-10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India.
| |
Collapse
|
2
|
Liu H, Wang S, Wang J, Guo X, Song Y, Fu K, Gao Z, Liu D, He W, Yang LL. Energy metabolism in health and diseases. Signal Transduct Target Ther 2025; 10:69. [PMID: 39966374 PMCID: PMC11836267 DOI: 10.1038/s41392-025-02141-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/08/2024] [Accepted: 12/25/2024] [Indexed: 02/20/2025] Open
Abstract
Energy metabolism is indispensable for sustaining physiological functions in living organisms and assumes a pivotal role across physiological and pathological conditions. This review provides an extensive overview of advancements in energy metabolism research, elucidating critical pathways such as glycolysis, oxidative phosphorylation, fatty acid metabolism, and amino acid metabolism, along with their intricate regulatory mechanisms. The homeostatic balance of these processes is crucial; however, in pathological states such as neurodegenerative diseases, autoimmune disorders, and cancer, extensive metabolic reprogramming occurs, resulting in impaired glucose metabolism and mitochondrial dysfunction, which accelerate disease progression. Recent investigations into key regulatory pathways, including mechanistic target of rapamycin, sirtuins, and adenosine monophosphate-activated protein kinase, have considerably deepened our understanding of metabolic dysregulation and opened new avenues for therapeutic innovation. Emerging technologies, such as fluorescent probes, nano-biomaterials, and metabolomic analyses, promise substantial improvements in diagnostic precision. This review critically examines recent advancements and ongoing challenges in metabolism research, emphasizing its potential for precision diagnostics and personalized therapeutic interventions. Future studies should prioritize unraveling the regulatory mechanisms of energy metabolism and the dynamics of intercellular energy interactions. Integrating cutting-edge gene-editing technologies and multi-omics approaches, the development of multi-target pharmaceuticals in synergy with existing therapies such as immunotherapy and dietary interventions could enhance therapeutic efficacy. Personalized metabolic analysis is indispensable for crafting tailored treatment protocols, ultimately providing more accurate medical solutions for patients. This review aims to deepen the understanding and improve the application of energy metabolism to drive innovative diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuo Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kun Fu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenjie Gao
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
3
|
Garg V, Ghay R, Goyal G, Saini RV. Exploring the Role of Acute Exercise-Induced Myokine Release in Glucose Metabolism and Insulin Sensitivity in Healthy and Diabetic Individuals. Cureus 2025; 17:e78991. [PMID: 40091956 PMCID: PMC11910891 DOI: 10.7759/cureus.78991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Background Exercise plays a significant role in influencing muscle metabolism and the secretion of myokines, which may have important therapeutic implications for managing type 2 diabetes mellitus (T2DM). This study explores the effects of a single session of moderate-intensity exercise on the levels of circulating myokines, specifically interleukin-6 (IL-6) and fractalkine, in individuals with T2DM compared to healthy controls. Methodology A total of 70 participants, including 35 individuals (50%) with T2DM and 35 healthy controls (50%) were enrolled in the study after taking their written informed consent. They took part in a 30-minute treadmill exercise session. Blood samples were collected before and after the exercise to measure fasting blood sugar (FBS), insulin, Homeostatic Model Assessment of Insulin Resistance (HOMA-IR), IL-6, and fractalkine levels, allowing for an assessment of the exercise's effects on both groups. Results In the post-exercise period, both groups demonstrated significant improvements in FBS, insulin levels, and HOMA-IR. Notably, IL-6 levels increased, while fractalkine levels decreased, indicating exercise's beneficial metabolic and anti-inflammatory effects. However, it is important to note that no significant correlation was observed between myokine levels and the markers of glucose metabolism. Conclusions This study demonstrates that acute exercise positively impacts glucose regulation and myokine modulation in both T2DM and healthy individuals. The findings support the inclusion of exercise as an effective strategy for improving metabolic health in diabetes management, highlighting the role of muscle-derived myokines in regulating glucose metabolism. Further research is needed to explore the long-term effects and mechanistic pathways involved.
Collapse
Affiliation(s)
- Vaishali Garg
- Physiology, Sri Guru Ram Das Institute of Medical Sciences and Research, Amritsar, IND
| | - Richa Ghay
- Physiology and Medical Education, Sri Guru Ram Das Institute of Medical Sciences and Research, Amritsar, IND
| | - Gurdev Goyal
- Physiology, Maharishi Markandeshwar (Deemed to be University), Ambala, IND
| | - Reena V Saini
- Biosciences and Technology, Maharishi Markandeshwar (Deemed to be University), Ambala, IND
| |
Collapse
|
4
|
Fiorenza M, Onslev J, Henríquez-Olguín C, Persson KW, Hesselager SA, Jensen TE, Wojtaszewski JFP, Hostrup M, Bangsbo J. Reducing the mitochondrial oxidative burden alleviates lipid-induced muscle insulin resistance in humans. SCIENCE ADVANCES 2024; 10:eadq4461. [PMID: 39475607 PMCID: PMC11524190 DOI: 10.1126/sciadv.adq4461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
Preclinical models suggest mitochondria-derived oxidative stress as an underlying cause of insulin resistance. However, it remains unknown whether this pathophysiological mechanism is conserved in humans. Here, we used an invasive in vivo mechanistic approach to interrogate muscle insulin action while selectively manipulating the mitochondrial redox state in humans. To this end, we conducted insulin clamp studies combining intravenous infusion of a lipid overload with intake of a mitochondria-targeted antioxidant (mitoquinone). Under lipid overload, selective modulation of mitochondrial redox state by mitoquinone enhanced insulin-stimulated glucose uptake in skeletal muscle. Mechanistically, mitoquinone did not affect canonical insulin signaling but augmented insulin-stimulated glucose transporter type 4 (GLUT4) translocation while reducing the mitochondrial oxidative burden under lipid oversupply. Complementary ex vivo studies in human muscle fibers exposed to high intracellular lipid levels revealed that mitoquinone improves features of mitochondrial bioenergetics, including diminished mitochondrial H2O2 emission. These findings provide translational and mechanistic evidence implicating mitochondrial oxidants in the development of lipid-induced muscle insulin resistance in humans.
Collapse
Affiliation(s)
- Matteo Fiorenza
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Johan Onslev
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Carlos Henríquez-Olguín
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago 1509, Chile
| | - Kaspar W. Persson
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Sofie A. Hesselager
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Thomas E. Jensen
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jørgen F. P. Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Morten Hostrup
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| | - Jens Bangsbo
- August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen 2100, Denmark
| |
Collapse
|
5
|
Okuma H, Tsuchiya K. Tissue-specific activation of insulin signaling as a potential target for obesity-related metabolic disorders. Pharmacol Ther 2024; 262:108699. [PMID: 39111411 DOI: 10.1016/j.pharmthera.2024.108699] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this "healthy adipose tissue expansion" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue.
Collapse
Affiliation(s)
- Hideyuki Okuma
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan
| | - Kyoichiro Tsuchiya
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan.
| |
Collapse
|
6
|
Chen K, Wang Y, Li D, Li J, Huang Y, Huang M, Ma H. Impact of diverse aerobic exercise plans on glycemic control, lipid levels, and functional activity in stroke patients with type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2024; 15:1389538. [PMID: 39359413 PMCID: PMC11446103 DOI: 10.3389/fendo.2024.1389538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Aims This study aimed to assess the effects of Low-to-Moderate Intensity Continuous Training (LMICT), Moderate-Intensity Interval Training (MIIT), and Reduced-Exertion High-Intensity Training (REHIT) on blood glucose regulation, functional recovery, and lipid levels in individuals who have experienced a stroke and are diagnosed with Type 2 Diabetes Mellitus (T2DM). Methods Forty-two T2DM stroke patients were randomly allocated to four groups: LMICT, MIIT, REHIT, and a control group (CON). Participants continuously monitored their blood glucose levels throughout the intervention using continuous glucose monitoring (CGM) devices. The study comprised two exercise intervention cycles: the first lasting from Day 3 to Day 14 and the second from Day 15 to Day 28, with the initial two days serving as contrasting periods. Primary outcomes encompassed CGM-derived blood glucose measurements, the Barthel Index (BI), Fugl-Meyer Assessment lower-extremity subscale (FMA-LE), and alterations in triglycerides (TG), total cholesterol (TC), high-density lipoprotein cholesterol (HDL-c), and low-density lipoprotein cholesterol (LDL-c). Results Compared with the CON, the MIIT group showed significant improvements in mean glucose (MG), glucose standard deviation (SD), time above range (TAR), and time in range (TIR). The REHIT group exhibited significantly reduced time below range (TBR), glucose SD, and coefficient of variation (CV). Regarding lipid levels, although the REHIT group achieved a significant reduction in TG levels compared with the CON, the overall effects of LMICT, MIIT, and REHIT on lipid profiles were relatively modest. Concerning functional recovery, the REHIT group significantly improved the BI and FMA-LE. Conclusion Although the short-term quantitative impact of exercise on lipid levels may be limited, both REHIT and MIIT significantly improved glycemic management and reduced glucose variability in post-stroke patients with Type 2 Diabetes Mellitus. Additionally, REHIT notably enhanced functional recovery.
Collapse
Affiliation(s)
- Kangcheng Chen
- School of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Yulong Wang
- Department of Rehabilitation, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Dongxia Li
- Department of Rehabilitation, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jun Li
- School of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Yong Huang
- Department of Rehabilitation, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Meiling Huang
- Department of Rehabilitation, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Haifeng Ma
- School of Athletic Performance, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
7
|
Nicolaisen TS, Sjøberg KA, Carl CS, Richter EA, Kiens B, Fritzen AM, Lundsgaard AM. Greater molecular potential for glucose metabolism in adipose tissue and skeletal muscle of women compared with men. FASEB J 2024; 38:e23845. [PMID: 39082199 DOI: 10.1096/fj.202302377r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 07/02/2024] [Accepted: 07/21/2024] [Indexed: 12/02/2024]
Abstract
Women typically have less muscle mass and more fat mass than men, while at the same time possessing similar or even greater whole-body insulin sensitivity. Our study aimed to investigate the molecular factors in primarily adipose tissue, but also in skeletal muscle, contributing to this sex difference. In healthy, moderately active premenopausal women and men with normal weight (28 ± 5 and 23 ± 3 years old; BMI 22.2 ± 1.9 and 23.7 ± 1.7) and in healthy, recreationally active women and men with overweight (32.2 ± 6 and 31.0 ± 5 years old; BMI 29.8 ± 4.3 & 30.9 ± 3.7) matched at age, BMI, and fitness level, we assessed insulin sensitivity and glucose tolerance with a hyperinsulinemic-euglycemic clamp or oral glucose tolerance test and studied subcutaneous adipose tissue and skeletal muscle samples with western blotting. Additionally, we traced glucose-stimulated glucose disposal in adipose tissues of female and male C57BL/6J littermate mice aged 16 weeks and measured glucose metabolic proteins. Our findings revealed greater protein expression related to glucose disposal in the subcutaneous adipose tissue (AKT2, insulin receptor, glucose transport 4) and skeletal muscle (hexokinase II, pyruvate dehydrogenase) in women compared to matched men with normal weight and with overweight. This increased protein capacity for glucose uptake extended to white adipose tissues of mice accompanied with ~2-fold greater glucose uptake compared to male mice. Furthermore, even in the obese state, women displayed better glucose tolerance than matched men, despite having 46% body fat and 20 kg less lean mass. In conclusion, our findings suggest that the superior potential for glucose disposal in female subcutaneous adipose tissue and skeletal muscle, driven by greater expression of various glucose metabolic proteins, compensates for their lower muscle mass. This likely explains women's superior glucose tolerance and tissue insulin sensitivity compared to men.
Collapse
Affiliation(s)
- Trine S Nicolaisen
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kim A Sjøberg
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Christian S Carl
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik A Richter
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Andreas M Fritzen
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Marie Lundsgaard
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk A/S, Søborg, Denmark
| |
Collapse
|
8
|
Annicchiarico A, Barile B, Buccoliero C, Nicchia GP, Brunetti G. Alternative therapeutic strategies in diabetes management. World J Diabetes 2024; 15:1142-1161. [PMID: 38983831 PMCID: PMC11229975 DOI: 10.4239/wjd.v15.i6.1142] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/17/2024] [Accepted: 04/12/2024] [Indexed: 06/11/2024] Open
Abstract
Diabetes is a heterogeneous metabolic disease characterized by elevated blood glucose levels resulting from the destruction or malfunction of pancreatic β cells, insulin resistance in peripheral tissues, or both, and results in a non-sufficient production of insulin. To adjust blood glucose levels, diabetic patients need exogenous insulin administration together with medical nutrition therapy and physical activity. With the aim of improving insulin availability in diabetic patients as well as ameliorating diabetes comorbidities, different strategies have been investigated. The first approaches included enhancing endogenous β cell activity or transplanting new islets. The protocol for this kind of intervention has recently been optimized, leading to standardized procedures. It is indicated for diabetic patients with severe hypoglycemia, complicated by impaired hypoglycemia awareness or exacerbated glycemic lability. Transplantation has been associated with improvement in all comorbidities associated with diabetes, quality of life, and survival. However, different trials are ongoing to further improve the beneficial effects of transplantation. Furthermore, to overcome some limitations associated with the availability of islets/pancreas, alternative therapeutic strategies are under evaluation, such as the use of mesenchymal stem cells (MSCs) or induced pluripotent stem cells for transplantation. The cotransplantation of MSCs with islets has been successful, thus providing protection against proinflammatory cytokines and hypoxia through different mechanisms, including exosome release. The use of induced pluripotent stem cells is recent and requires further investigation. The advantages of MSC implantation have also included the improvement of diabetes-related comorbidities, such as wound healing. Despite the number of advantages of the direct injection of MSCs, new strategies involving biomaterials and scaffolds have been developed to improve the efficacy of mesenchymal cell delivery with promising results. In conclusion, this paper offered an overview of new alternative strategies for diabetes management while highlighting some limitations that will need to be overcome by future approaches.
Collapse
Affiliation(s)
- Alessia Annicchiarico
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Cinzia Buccoliero
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| |
Collapse
|
9
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
10
|
Logesh R, Hari B, Chidambaram K, Das N. Molecular effects of Vitamin-D and PUFAs metabolism in skeletal muscle combating Type-II diabetes mellitus. Gene 2024; 904:148216. [PMID: 38307219 DOI: 10.1016/j.gene.2024.148216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 01/10/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024]
Abstract
Multiple post-receptor intracellular alterations such as impaired glucose transfer, glucose phosphorylation, decreased glucose oxidation, and glycogen production contribute to insulin resistance (IR) in skeletal muscle, manifested by diminished insulin-stimulated glucose uptake. Type-2 diabetes mellites (T2DM) has caused by IR, which is also seen in obese patients and those with metabolic syndrome. The Vitamin-D receptor (VDR) and poly unsaturated fatty acids (PUFAs) roles in skeletal muscle growth, shapes, and function for combating type-2 diabetes have been clarified throughout this research. VDR and PUFAs appears to show a variety of effects on skeletal muscle, in addition it shows a promising role on bone and mineral homeostasis. Individuals having T2DM are reported to suffer from severe muscular weakness and alterations in shape of the muscle. Several studies have investigated the effect on VDR on muscular strength and mass, which leads to Vitamin-D deficiency (VDD) in individuals, in which most commonly seen in elderly. VDR has been shown to affect skeletal cellular proliferation, intracellular calcium handling, as well as genomic activity in a variety of different ways such as muscle metabolism, insulin sensitivity, which is the major characteristic pathogenesis for IR in combating T2DM. The identified VDR gene polymorphisms are ApaI, TaqI, FokI, and BsmI that are associated with T2DM. This review collates informations on the mechanisms by which VDR activation takes place in skeletal muscles. Despite the significant breakthroughs made in recent decades, various studies show that IR affects VDR and PUFAs metabolism in skeletal muscle. Therefore, this review collates the data to show the role of VDR and PUFAs in the skeletal muscles to combat T2DM.
Collapse
Affiliation(s)
- Rajan Logesh
- Department of Pharmacognosy, JSS College of Pharmacy, Mysuru, JSS Academy of Higher Education & Research, Karnataka, India.
| | - Balaji Hari
- TIFAC CORE in Herbal Drugs, Department of Pharmacognosy, JSS Academy of Higher Education & Research, JSS College of Pharmacy, The Nilgiris, Ooty 643001, Tamil Nadu, India
| | - Kumarappan Chidambaram
- Department of Pharmacology, College of Pharmacy, King Khalid University, Al-Qara, Asir Province, Saudi Arabia
| | - Niranjan Das
- Department of Chemistry, Iswar Chandra Vidyasagar College, Belonia 799155, Tripura, India
| |
Collapse
|
11
|
Knudsen CB, Nielsen J, Ørtenblad N, Mohr M, Overgaard K, Vigh-Larsen JF. No net utilization of intramuscular lipid droplets during repeated high-intensity intermittent exercise. Am J Physiol Endocrinol Metab 2023; 325:E700-E710. [PMID: 37877795 DOI: 10.1152/ajpendo.00298.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/26/2023]
Abstract
Intramuscular lipids are stored as subsarcolemmal or intramyofibrillar droplets with potential diverse roles in energy metabolism. We examined intramuscular lipid utilization through transmission electron microscopy during repeated high-intensity intermittent exercise, an aspect that is hitherto unexplored. Seventeen moderately to well-trained males underwent three periods (EX1-EX3) of 10 × 45-s high-intensity cycling [∼100%-120% Wattmax (Wmax)] combined with maximal repeated sprints (∼250%-300% Wmax). M. vastus lateralis biopsies were obtained at baseline, after EX1, and EX3. During the complete exercise session, no net decline in either subsarcolemmal or intermyofibrillar lipid volume density occurred. However, a temporal relationship emerged for subsarcolemmal lipids with an ∼11% increase in droplet size after EX1 (P = 0.024), which reverted to baseline levels after EX3 accompanied by an ∼30% reduction in the numerical density of subsarcolemmal lipid droplets compared with both baseline (P = 0.019) and after EX1 (P = 0.018). Baseline distinctions were demonstrated with an approximately twofold higher intermyofibrillar lipid volume in type 1 versus type 2 fibers (P = 0.008), mediated solely by a higher number rather than the size of lipid droplets (P < 0.001). No fiber-type-specific differences were observed in subsarcolemmal lipid volume although type 2 fibers exhibited ∼17% larger droplets (P = 0.034) but a lower numerical density (main effect; P = 0.010) including 3% less droplets at baseline. Collectively, these findings suggest that intramuscular lipids do not serve as an important substrate during high-intensity intermittent exercise; however, the repeated exercise pattern mediated a temporal remodeling of the subsarcolemmal lipid pool. Furthermore, fiber-type- and compartment-specific differences were found at baseline underscoring the heterogeneity in lipid droplet deposition.NEW & NOTEWORTHY Undertaking a severe repeated high-intensity intermittent exercise protocol led to no net decline in neither subsarcolemmal nor intermyofibrillar lipid content in the thigh muscle of young moderately to well-trained participants. However, a temporal remodeling of the subsarcolemmal pool of lipid droplets did occur indicative of potential transient lipid accumulation. Moreover, baseline fiber-type distinctions in subcellular lipid droplet deposition were present underscoring the diversity in lipid droplet storage among fiber types and subcellular regions.
Collapse
Affiliation(s)
- Christian B Knudsen
- Department of Public Health, Research Unit for Exercise Biology, Aarhus University, Aarhus, Denmark
| | - Joachim Nielsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Niels Ørtenblad
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| | - Magni Mohr
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- Centre of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Kristian Overgaard
- Department of Public Health, Research Unit for Exercise Biology, Aarhus University, Aarhus, Denmark
| | - Jeppe F Vigh-Larsen
- Department of Public Health, Research Unit for Exercise Biology, Aarhus University, Aarhus, Denmark
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
12
|
Peeters WM, Gram M, Dias GJ, Vissers MCM, Hampton MB, Dickerhof N, Bekhit AE, Black MJ, Oxbøll J, Bayer S, Dickens M, Vitzel K, Sheard PW, Danielson KM, Hodges LD, Brønd JC, Bond J, Perry BG, Stoner L, Cornwall J, Rowlands DS. Changes to insulin sensitivity in glucose clearance systems and redox following dietary supplementation with a novel cysteine-rich protein: A pilot randomized controlled trial in humans with type-2 diabetes. Redox Biol 2023; 67:102918. [PMID: 37812879 PMCID: PMC10570009 DOI: 10.1016/j.redox.2023.102918] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023] Open
Abstract
We recently developed a novel keratin-derived protein (KDP) rich in cysteine, glycine, and arginine, with the potential to alter tissue redox status and insulin sensitivity. The KDP was tested in 35 human adults with type-2 diabetes mellitus (T2DM) in a 14-wk randomised controlled pilot trial comprising three 2×20 g supplemental protein/day arms: KDP-whey (KDPWHE), whey (WHEY), non-protein isocaloric control (CON), with standardised exercise. Outcomes were measured morning fasted and following insulin-stimulation (80 mU/m2/min hyperinsulinaemic-isoglycaemic clamp). With KDPWHE supplementation there was good and very-good evidence for moderate-sized increases in insulin-stimulated glucose clearance rate (GCR; 26%; 90% confidence limits, CL 2%, 49%) and skeletal-muscle microvascular blood flow (46%; 16%, 83%), respectively, and good evidence for increased insulin-stimulated sarcoplasmic GLUT4 translocation (18%; 0%, 39%) vs CON. In contrast, WHEY did not effect GCR (-2%; -25%, 21%) and attenuated HbA1c lowering (14%; 5%, 24%) vs CON. KDPWHE effects on basal glutathione in erythrocytes and skeletal muscle were unclear, but in muscle there was very-good evidence for large increases in oxidised peroxiredoxin isoform 2 (oxiPRX2) (19%; 2.2%, 35%) and good evidence for lower GPx1 concentrations (-40%; -4.3%, -63%) vs CON; insulin stimulation, however, attenuated the basal oxiPRX2 response (4%; -16%, 24%), and increased GPx1 (39%; -5%, 101%) and SOD1 (26%; -3%, 60%) protein expression. Effects of KDPWHE on oxiPRX3 and NRF2 content, phosphorylation of capillary eNOS and insulin-signalling proteins upstream of GLUT4 translocation AktSer437 and AS160Thr642 were inconclusive, but there was good evidence for increased IRSSer312 (41%; 3%, 95%), insulin-stimulated NFκB-DNA binding (46%; 3.4%, 105%), and basal PAK-1Thr423/2Thr402 phosphorylation (143%; 66%, 257%) vs WHEY. Our findings provide good evidence to suggest that dietary supplementation with a novel edible keratin protein in humans with T2DM may increase glucose clearance and modify skeletal-muscle tissue redox and insulin sensitivity within systems involving peroxiredoxins, antioxidant expression, and glucose uptake.
Collapse
Affiliation(s)
- W M Peeters
- Metabolic and Microvascular Laboratory, School of Sport, Exercise and Nutrition, Massey University, Wellington, Auckland, New Zealand; School of Biomedical, Nutritional and Sport Science, Newcastle University, United Kingdom
| | - M Gram
- Metabolic and Microvascular Laboratory, School of Sport, Exercise and Nutrition, Massey University, Wellington, Auckland, New Zealand
| | - G J Dias
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - M C M Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - M B Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - N Dickerhof
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - A E Bekhit
- Department of Food Sciences, University of Otago, Dunedin, New Zealand
| | - M J Black
- Metabolic and Microvascular Laboratory, School of Sport, Exercise and Nutrition, Massey University, Wellington, Auckland, New Zealand
| | - J Oxbøll
- Metabolic and Microvascular Laboratory, School of Sport, Exercise and Nutrition, Massey University, Wellington, Auckland, New Zealand
| | - S Bayer
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - M Dickens
- School of Health Sciences, Massey University, Wellington, Auckland, New Zealand
| | - K Vitzel
- School of Health Sciences, Massey University, Wellington, Auckland, New Zealand
| | - P W Sheard
- Department of Physiology, University of Otago, Dunedin, New Zealand
| | - K M Danielson
- Department of Anaesthesiology and Surgery, University of Otago, Wellington, New Zealand
| | - L D Hodges
- Metabolic and Microvascular Laboratory, School of Sport, Exercise and Nutrition, Massey University, Wellington, Auckland, New Zealand
| | - J C Brønd
- Department of Sports Science and Clinical Biomechanics, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - J Bond
- Metabolic and Microvascular Laboratory, School of Sport, Exercise and Nutrition, Massey University, Wellington, Auckland, New Zealand
| | - B G Perry
- School of Health Sciences, Massey University, Wellington, Auckland, New Zealand
| | - L Stoner
- Department of Exercise and Sport Science, University of North Carolina, Chapel Hill, USA
| | - J Cornwall
- Centre for Early Learning in Medicine, University of Otago, Dunedin, New Zealand
| | - D S Rowlands
- Metabolic and Microvascular Laboratory, School of Sport, Exercise and Nutrition, Massey University, Wellington, Auckland, New Zealand.
| |
Collapse
|
13
|
Alharbi A, Li J, Womack E, Farrow M, Yarar-Fisher C. The Effect of Lower Limb Combined Neuromuscular Electrical Stimulation on Skeletal Muscle Signaling for Glucose Utilization, Myofiber Distribution, and Metabolic Function after Spinal Cord Injury. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6958. [PMID: 37887696 PMCID: PMC10606374 DOI: 10.3390/ijerph20206958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/29/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023]
Abstract
Maintaining healthy myofiber type and metabolic function early after spinal cord injury (SCI) may prevent chronic metabolic disorders. This study compares the effects of a 2-5 week combined (aerobic + resistance) neuromuscular electrical stimulation (Comb-NMES) regimen versus a sham control treatment on muscle protein signaling for glucose uptake, myofiber type distribution, and metabolic function. Twenty participants (31 ± 9 years of age) with an SCI (C4-L1, AIS level A-C) within 14 days of the SCI were randomly assigned to control (N = 8) or Comb-NMES (N = 12). Sessions were given three times per week. Fasting blood samples and vastus lateralis muscle biopsies were collected 24-48 h before or after the last session. Western blots were performed to quantify proteins, immunohistochemical analyses determined muscle myofiber distribution, and enzymatic assays were performed to measure serum glucose, insulin, and lipids. Our main findings include a decrease in fasting glucose (p < 0.05) and LDL-C (p < 0.05) levels, an upregulation of CamKII and Hexokinase (p < 0.05), and an increase in type I (+9%) and a decrease in type IIx (-36%) myofiber distribution in response to Comb-NMES. Our findings suggest that maintaining healthy myofiber type and metabolic function may be achieved via early utilization of Comb-NMES.
Collapse
Affiliation(s)
- Amal Alharbi
- Department of Physical Therapy, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Jia Li
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH 43210, USA; (J.L.); (M.F.)
| | - Erika Womack
- Department of Biochemistry, Molecular Biology, Entomology and Plant Pathology, Mississippi State University, Starkville, MS 39762, USA;
| | - Matthew Farrow
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH 43210, USA; (J.L.); (M.F.)
| | - Ceren Yarar-Fisher
- Department of Physical Medicine and Rehabilitation, Ohio State University, Columbus, OH 43210, USA; (J.L.); (M.F.)
- Department of Neuroscience, Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
14
|
Chong HW, Son J, Chae C, Jae C. The relationship between skeletal muscle mass and the KOSHA cardiovascular risk in obese male workers. Ann Occup Environ Med 2023; 35:e40. [PMID: 38029272 PMCID: PMC10654537 DOI: 10.35371/aoem.2023.35.e40] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/08/2023] [Accepted: 09/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background Efforts for the prevention and management of cardiovascular diseases (CVDs) in workers have been actively pursued. Obesity is one of the important risk factors related to CVDs. Obesity has various metabolic characteristics, and some individuals can be metabolically healthy. Body composition including skeletal muscle mass is known to have protective effect in obesity. The study aims to investigate the association between skeletal muscle mass and Korea Occupational Safety and Health Agency (KOSHA) CVD risk among obese male manufacturing workers in Korea and to identify appropriate indicators of skeletal muscle mass for predicting risk of CVDs. Methods The study was conducted on 2,007 obese male workers at a manufacturing industry aged more than 19 years. Skeletal muscle mass, skeletal muscle index (SMI), skeletal muscle mass percent (SMM%) and skeletal muscle to body fat ratio (MFR) were used to evaluate body composition and these indicators were divided into quartiles. The odds ratios (ORs) and 95% confidence intervals (CIs) for the KOSHA CVD risk groups according to quartiles of skeletal muscle mass indicators were estimated using ordinal logistic regression analysis. Results The OR for the KOSHA CVD risk groups in the highest quartile of SMI was 1.67 (95% CI: 1.42-1.92), while the ORs for the KOSHA CVD risk groups in the highest quartiles of SMM%, SMM/body mass index (BMI), and MFR were 0.47 (95% CI: 0.22-0.72), 0.51 (95% CI: 0.05-0.76), and 0.48 (95% CI: 0.23-0.74), respectively. Conclusions We found that high SMI increase the likelihood of high risk of CVDs, while high SMM%, SMM/BMI, and MFR lower the likelihood of high risk of CVDs. Accurate evaluation of skeletal muscle mass can help assess the cardiovascular risk in obese male workers.
Collapse
Affiliation(s)
- Hyo Won Chong
- Department of Occupational and Environmental Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - JunSeok Son
- Department of Occupational and Environmental Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Changho Chae
- Department of Occupational and Environmental Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Changho Jae
- Department of Occupational and Environmental Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| |
Collapse
|
15
|
Macedo ACPD, Schaan CW, Bock PM, Pinto MBD, Botton CE, Umpierre D, Schaan BD. Cardiorespiratory fitness in individuals with type 2 diabetes mellitus: a systematic review and meta-analysis. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:e230040. [PMID: 37738467 PMCID: PMC10665050 DOI: 10.20945/2359-4292-2023-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 05/30/2023] [Indexed: 09/24/2023]
Abstract
Objective To conduct a systematic review and meta-analysis assessing the cardiorespiratory fitness (CRF) among individuals with and without type 2 diabetes. Materials and methods The current review was registered in PROSPERO under the number CRD42018082718. MEDLINE, EMBASE, and Cochrane Library databases were searched from inception through February 2022. Eligibility criteria consisted of observational or interventional studies that evaluated CRF through cardiopulmonary exercise testing or six-minute walk test in individuals with type 2 diabetes compared with individuals without type 2 diabetes. For data extraction, we used baseline CRF assessments of randomized clinical trials or follow-up CRF assessments in observational studies. We performed a meta-analysis using maximal oxygen consumption (VO2 max), and distance walked in the 6MWT as primary outcomes. They were extracted and expressed as mean differences (MDs) and 95% CIs between treatment and comparator groups. The meta-analysis was conducted using Review Manager (RevMan) software. Results Out of 8,347 studies retrieved, 77 were included. Compared with individuals without type 2 diabetes, individuals with diabetes achieved a lower VO2 max (-5.84 mL.kg-1.min-1, 95% CI -6.93, -4.76 mL.kg-1.min-1, p = <0.0001; I2 = 91%, p for heterogeneity < 0.0001), and a smaller distance walked in 6MWT (-93.30 meters, 95% CI -141.2, -45.4 meters, p > 0.0001; I2: 94%, p for heterogeneity < 0.0001). Conclusion Type 2 diabetes was associated with lower cardiorespiratory fitness, as observed by lower VO2 max on maximal tests, and smaller distance walked in 6MWT, however the quality of studies was low.
Collapse
Affiliation(s)
- Aline Chagastelles Pinto de Macedo
- Universidade Federal do Rio Grande do Sul, Programa de Pós-graduaçÃo em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Camila Wohlgemuth Schaan
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Patricia Martins Bock
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil,
- Faculdades Integradas de Taquara, Taquara, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Mariana Brutto de Pinto
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Cintia Ehlers Botton
- Instituto de AvaliaçÃo de Tecnologia em Saúde (IATS) - CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Ceará, Instituto de EducaçÃo Física e Esportes, Fortaleza, CE, Brasil
- Programa de Mestrado em Fisioterapia e Funcionalidade, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - Daniel Umpierre
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Instituto de AvaliaçÃo de Tecnologia em Saúde (IATS) - CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Beatriz D Schaan
- Universidade Federal do Rio Grande do Sul, Programa de Pós-graduaçÃo em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Instituto de AvaliaçÃo de Tecnologia em Saúde (IATS) - CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| |
Collapse
|
16
|
Lloyd EM, Pinniger GJ, Murphy RM, Grounds MD. Slow or fast: Implications of myofibre type and associated differences for manifestation of neuromuscular disorders. Acta Physiol (Oxf) 2023; 238:e14012. [PMID: 37306196 DOI: 10.1111/apha.14012] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Many neuromuscular disorders can have a differential impact on a specific myofibre type, forming the central premise of this review. The many different skeletal muscles in mammals contain a spectrum of slow- to fast-twitch myofibres with varying levels of protein isoforms that determine their distinctive contractile, metabolic, and other properties. The variations in functional properties across the range of classic 'slow' to 'fast' myofibres are outlined, combined with exemplars of the predominantly slow-twitch soleus and fast-twitch extensor digitorum longus muscles, species comparisons, and techniques used to study these properties. Other intrinsic and extrinsic differences are discussed in the context of slow and fast myofibres. These include inherent susceptibility to damage, myonecrosis, and regeneration, plus extrinsic nerves, extracellular matrix, and vasculature, examined in the context of growth, ageing, metabolic syndrome, and sexual dimorphism. These many differences emphasise the importance of carefully considering the influence of myofibre-type composition on manifestation of various neuromuscular disorders across the lifespan for both sexes. Equally, understanding the different responses of slow and fast myofibres due to intrinsic and extrinsic factors can provide deep insight into the precise molecular mechanisms that initiate and exacerbate various neuromuscular disorders. This focus on the influence of different myofibre types is of fundamental importance to enhance translation for clinical management and therapies for many skeletal muscle disorders.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
17
|
Møller LLV, Ali MS, Davey J, Raun SH, Andersen NR, Long JZ, Qian H, Jeppesen JF, Henriquez-Olguin C, Frank E, Jensen TE, Højlund K, Wojtaszewski JFP, Nielsen J, Chiu TT, Jedrychowski MP, Gregorevic P, Klip A, Richter EA, Sylow L. The Rho guanine dissociation inhibitor α inhibits skeletal muscle Rac1 activity and insulin action. Proc Natl Acad Sci U S A 2023; 120:e2211041120. [PMID: 37364105 PMCID: PMC10318982 DOI: 10.1073/pnas.2211041120] [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: 07/11/2022] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
The molecular events governing skeletal muscle glucose uptake have pharmacological potential for managing insulin resistance in conditions such as obesity, diabetes, and cancer. With no current pharmacological treatments to target skeletal muscle insulin sensitivity, there is an unmet need to identify the molecular mechanisms that control insulin sensitivity in skeletal muscle. Here, the Rho guanine dissociation inhibitor α (RhoGDIα) is identified as a point of control in the regulation of insulin sensitivity. In skeletal muscle cells, RhoGDIα interacted with, and thereby inhibited, the Rho GTPase Rac1. In response to insulin, RhoGDIα was phosphorylated at S101 and Rac1 dissociated from RhoGDIα to facilitate skeletal muscle GLUT4 translocation. Accordingly, siRNA-mediated RhoGDIα depletion increased Rac1 activity and elevated GLUT4 translocation. Consistent with RhoGDIα's inhibitory effect, rAAV-mediated RhoGDIα overexpression in mouse muscle decreased insulin-stimulated glucose uptake and was detrimental to whole-body glucose tolerance. Aligning with RhoGDIα's negative role in insulin sensitivity, RhoGDIα protein content was elevated in skeletal muscle from insulin-resistant patients with type 2 diabetes. These data identify RhoGDIα as a clinically relevant controller of skeletal muscle insulin sensitivity and whole-body glucose homeostasis, mechanistically by modulating Rac1 activity.
Collapse
Affiliation(s)
- Lisbeth L. V. Møller
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Mona S. Ali
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Jonathan Davey
- The Centre for Muscle Research, Department of Physiology, The University of Melbourne, Parkville, VIC3010, Australia
| | - Steffen H. Raun
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Nicoline R. Andersen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Jonathan Z. Long
- Department of Pathology, Stanford University School of Medicine and Stanford, Stanford University, Stanford, CA94305
| | - Hongwei Qian
- The Centre for Muscle Research, Department of Physiology, The University of Melbourne, Parkville, VIC3010, Australia
| | - Jacob F. Jeppesen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Carlos Henriquez-Olguin
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, 7501015Santiago, Chile
| | - Emma Frank
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Thomas E. Jensen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, 5000Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000Odense C, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5000Odense C, Denmark
| | - Jørgen F. P. Wojtaszewski
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Joachim Nielsen
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, 5230Odense M, Denmark
| | - Tim T. Chiu
- Cell Biology Program, The Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A1, Canada
- Department of Physiology, University of Toronto, Toronto, ONM5S 1A1, Canada
- Department of Paediatrics, University of Toronto, Toronto, ONM5S 1A1, Canada
| | - Mark P. Jedrychowski
- Department of Cell Biology, Harvard Medical School, Boston, MA02115
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Paul Gregorevic
- The Centre for Muscle Research, Department of Physiology, The University of Melbourne, Parkville, VIC3010, Australia
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, ONM5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ONM5S 1A1, Canada
- Department of Physiology, University of Toronto, Toronto, ONM5S 1A1, Canada
- Department of Paediatrics, University of Toronto, Toronto, ONM5S 1A1, Canada
| | - Erik A. Richter
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
| | - Lykke Sylow
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, 2200Copenhagen N, Denmark
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, 2200Copenhagen N, Denmark
| |
Collapse
|
18
|
Kim CJ, Singh C, Lee C, DiMagno K, O'Donnell M, Kaczmarek M, Ahmed A, Salvo‐Schaich J, Perez A, Letsou W, Sepulveda MC, Ramos RL, Hadjiargyrou M. Mustn1 ablation in skeletal muscle results in increased glucose tolerance concomitant with upregulated GLUT expression in male mice. Physiol Rep 2023; 11:e15674. [PMID: 37170065 PMCID: PMC10175242 DOI: 10.14814/phy2.15674] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 05/13/2023] Open
Abstract
Glucose homeostasis is closely regulated to maintain energy requirements of vital organs and skeletal muscle plays a crucial role in this process. Mustn1 is expressed during embryonic and postnatal skeletal muscle development and its function has been implicated in myogenic differentiation and myofusion. Whether Mustn1 plays a role in glucose homeostasis in anyway remains largely unknown. As such, we deleted Mustn1 in skeletal muscle using a conditional knockout (KO) mouse approach. KO mice did not reveal any specific gross phenotypic alterations in skeletal muscle. However, intraperitoneal glucose tolerance testing (IPGTT) revealed that 2-month-old male KO mice had significantly lower glycemia than their littermate wild type (WT) controls. These findings coincided with mRNA changes in genes known to be involved in glucose metabolism, tolerance, and insulin sensitivity; 2-month-old male KO mice had significantly higher expression of GLUT1 and GLUT10 transporters, MUP-1 while OSTN expression was lower. These differences in glycemia and gene expression were statistically insignificant after 4 months. Identical experiments in female KO and WT control mice did not indicate any differences at any age. Our results suggest a link between Mustn1 expression and glucose homeostasis during a restricted period of skeletal muscle development/maturation. While this is an observational study, Mustn1's relationship to glucose homeostasis appears to be more complex with a possible connection to other key proteins such as GLUTs, MUP-1, and OSTN. Additionally, our data indicate temporal and sex differences. Lastly, our findings strengthen the notion that Mustn1 plays a role in the metabolic capacity of skeletal muscle.
Collapse
Affiliation(s)
- Charles J. Kim
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
- Department of Biological and Chemical SciencesNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Chanpreet Singh
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Christine Lee
- Department of Biological and Chemical SciencesNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Kevin DiMagno
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Madison O'Donnell
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Marina Kaczmarek
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Arhum Ahmed
- Department of Biological and Chemical SciencesNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Jessica Salvo‐Schaich
- Department of Biological and Chemical SciencesNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Alexis Perez
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - William Letsou
- Department of Biological and Chemical SciencesNew York Institute of TechnologyOld WestburyNew YorkUSA
| | | | - Raddy L. Ramos
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
| | - Michael Hadjiargyrou
- College of Osteopathic MedicineNew York Institute of TechnologyOld WestburyNew YorkUSA
- Department of Biological and Chemical SciencesNew York Institute of TechnologyOld WestburyNew YorkUSA
| |
Collapse
|
19
|
Chen X, Tong YL, Ren ZM, Chen SS, Mei XY, Zhou QY, Dai GH. Hypoglycemic mechanisms of Polygonatum sibiricum polysaccharide in db/db mice via regulation of glycolysis/gluconeogenesis pathway and alteration of gut microbiota. Heliyon 2023; 9:e15484. [PMID: 37128343 PMCID: PMC10147986 DOI: 10.1016/j.heliyon.2023.e15484] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/02/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023] Open
Abstract
Polygonatum rhizoma polysaccharide (PP) is a main ingredient of Polygonatum rhizoma , which is both food and traditional herbal medicine. In this study, we aimed to investigate the hypoglycemic effect of PP and the underlying mechanisms in db/db mice. Our finding showed that PP significantly ameliorates diabetic symptoms by reducing glucose levels in blood and urine and increasing insulin and leptin abundance in the serum. Histopathological examination revealed that PP improved the pathological state and increased hepatic glycogen storage in liver. Additionally, RT-qPCR results indicated that PP significantly down-regulated the expression of phosphoenolpyruvate carboxykinase 1. Furthermore, 16s rRNA sequencing results demonstrated that PP intervention resulted in an increase in beneficial bacteria genus and a reduction in harmful genus. Redundancy analysis revealed the correlation between intestinal flora and clinical factors. Taken together, these results suggest that PP has a significant hypoglycemic effect on type 2 diabetes (T2D) through up-regulating serum insulin and leptin, as well as hepatic glycogen storage, and down-regulating hepatic phosphoenolpyruvate carboxykinase 1 expression, as well as modulating gut microbiota composition. In conclusion, this study investigated the mechanisms of PP in the treatment of diabetes in db/db mice. To the best of our knowledge, this is the first study to explore the positive and negative correlations between gut microbiota and clinical factors, such as oxidative stress injury in liver and glucose related indicators in the blood.
Collapse
Affiliation(s)
- Xuan Chen
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Ye-ling Tong
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Ze-ming Ren
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Si-si Chen
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Xi-yu Mei
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Qing-yun Zhou
- Second Clinical Medical School, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, People's Republic of China
| | - Guan-hai Dai
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
- Corresponding author.
| |
Collapse
|
20
|
San-Millán I. The Key Role of Mitochondrial Function in Health and Disease. Antioxidants (Basel) 2023; 12:antiox12040782. [PMID: 37107158 PMCID: PMC10135185 DOI: 10.3390/antiox12040782] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
The role of mitochondrial function in health and disease has become increasingly recognized, particularly in the last two decades. Mitochondrial dysfunction as well as disruptions of cellular bioenergetics have been shown to be ubiquitous in some of the most prevalent diseases in our society, such as type 2 diabetes, cardiovascular disease, metabolic syndrome, cancer, and Alzheimer's disease. However, the etiology and pathogenesis of mitochondrial dysfunction in multiple diseases have yet to be elucidated, making it one of the most significant medical challenges in our history. However, the rapid advances in our knowledge of cellular metabolism coupled with the novel understanding at the molecular and genetic levels show tremendous promise to one day elucidate the mysteries of this ancient organelle in order to treat it therapeutically when needed. Mitochondrial DNA mutations, infections, aging, and a lack of physical activity have been identified to be major players in mitochondrial dysfunction in multiple diseases. This review examines the complexities of mitochondrial function, whose ancient incorporation into eukaryotic cells for energy purposes was key for the survival and creation of new species. Among these complexities, the tightly intertwined bioenergetics derived from the combustion of alimentary substrates and oxygen are necessary for cellular homeostasis, including the production of reactive oxygen species. This review discusses different etiological mechanisms by which mitochondria could become dysregulated, determining the fate of multiple tissues and organs and being a protagonist in the pathogenesis of many non-communicable diseases. Finally, physical activity is a canonical evolutionary characteristic of humans that remains embedded in our genes. The normalization of a lack of physical activity in our modern society has led to the perception that exercise is an "intervention". However, physical activity remains the modus vivendi engrained in our genes and being sedentary has been the real intervention and collateral effect of modern societies. It is well known that a lack of physical activity leads to mitochondrial dysfunction and, hence, it probably becomes a major etiological factor of many non-communicable diseases affecting modern societies. Since physical activity remains the only stimulus we know that can improve and maintain mitochondrial function, a significant emphasis on exercise promotion should be imperative in order to prevent multiple diseases. Finally, in populations with chronic diseases where mitochondrial dysfunction is involved, an individualized exercise prescription should be crucial for the "metabolic rehabilitation" of many patients. From lessons learned from elite athletes (the perfect human machines), it is possible to translate and apply multiple concepts to the betterment of populations with chronic diseases.
Collapse
Affiliation(s)
- Iñigo San-Millán
- Department of Human Physiology and Nutrition, University of Colorado, Colorado Springs, CO 80198, USA
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
21
|
Abstract
The global prevalences of obesity and type 2 diabetes mellitus have reached epidemic status, presenting a heavy burden on society. It is therefore essential to find novel mechanisms and targets that could be utilized in potential treatment strategies and, as such, intracellular membrane trafficking has re-emerged as a regulatory tool for controlling metabolic homeostasis. Membrane trafficking is an essential physiological process that is responsible for the sorting and distribution of signalling receptors, membrane transporters and hormones or other ligands between different intracellular compartments and the plasma membrane. Dysregulation of intracellular transport is associated with many human diseases, including cancer, neurodegeneration, immune deficiencies and metabolic diseases, such as type 2 diabetes mellitus and its associated complications. This Review focuses on the latest advances on the role of endosomal membrane trafficking in metabolic physiology and pathology in vivo, highlighting the importance of this research field in targeting metabolic diseases.
Collapse
Affiliation(s)
- Jerome Gilleron
- Université Côte d'Azur, Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1065 C3M, Team Cellular and Molecular Pathophysiology of Obesity, Nice, France.
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
22
|
Mezghani N, Ammar A, Boukhris O, Abid R, Hadadi A, Alzahrani TM, Trabelsi O, Boujelbane MA, Masmoudi L, Ouergui I, Jamoussi K, Mnif M, Mejdoub H, Zmijewski P, Glenn JM, Trabelsi K, Chtourou H. The Impact of Exercise Training Intensity on Physiological Adaptations and Insulin Resistance in Women with Abdominal Obesity. Healthcare (Basel) 2022; 10:2533. [PMID: 36554057 PMCID: PMC9778339 DOI: 10.3390/healthcare10122533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Abdominal obesity has emerged globally as a major public health issue due to its high prevalence and morbidity. The benefits of physical exercise among the obese population are well documented. However, the optimal exercise intensity for reducing body fat and preventing insulin resistance and metabolic disorders is still under debate. This study aimed to examine the effects of three different intensities of combined endurance and strength training programs on anthropometric variables, physiological and muscular adaptations, and insulin sensitivity. Forty-three obese young women (age 26.4 ± 4.7 years, BMI 33.1 ± 2.5 kg/m2) were randomly assigned to one of four groups: a control group (G0), a moderate-intensity training group (G50, exercising brisk walking at 50% heart rate reserve HRR), a high-intensity training group (G75, exercise jogging at 75% HRR), and an alternated-intensity training group (G50/75, exercise brisk-walking/jogging at 50−75% HRR) with additional strength training once a week for each group. Body composition, waist circumference (WC), fasting blood glucose, insulin sensitivity and resistance (Homa-IR), resting heart rate (RHR), 6-min walk distance (6MWD), 1-repetition maximum (1-RM), and time to exhaustion (TTE) at 45% and 75% maximal voluntary contraction (MVC) for both the flexor and extensor muscle groups of the knees, were recorded before and after three months of exercise training. All training groups showed significant decreases in body mass, BMI, total body fat, body fat percentage, WC, abdominal and visceral mass (p < 0.001), with a greater reduction of body mass and BMI in G75 (p < 0.05). Lean mass increased significantly only in G50/75 (p < 0.05). The insulin sensitivity and Homa-IR decreased in the three training groups (p < 0.01), with greater enhanced resistance in G50 compared to G75 and G50/75 (p < 0.05). In contrast, there were no pre-post changes in all groups for fasting blood glucose (p > 0.05). 1-RM and TTE of the knee flexor and extensor muscles were improved in the three groups (p < 0.01), with greater improvement in G50/75 for 1RM and G75 in most of the TTE parameters (p < 0.05). RHR decreased and 6MWD increased significantly in the three training groups (p < 0.01), with greater 6MWD improvement in G75 (p < 0.05). In conclusion, the three training intensities seem to generate benefits in terms of body composition, physiological and muscular adaptations, and insulin resistance. High training intensity resulted in greater improvements in body mass, BMI, and endurance and strength, whereas moderate training intensity resulted in greater improvements of insulin resistance and homo-IR. Following alternate-intensity training, greater improvements were observed in lean mass and maximal strength performance.
Collapse
Affiliation(s)
- Nourhen Mezghani
- Department of Sport Sciences, College of Education, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Achraf Ammar
- Department of Training and Movement Science, Institute of Sport Science, Johannes Gutenberg-University Mainz, 55099 Mainz, Germany
- Interdisciplinary Laboratory in Neurosciences, Physiology and Psychology: Physical Activity, Health and Learning (LINP2), UFR STAPS, UPL, Paris Nanterre University, 39200 Nanterre, France
- High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
| | - Omar Boukhris
- High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
- Physical Activity, Sport, and Health, UR18JS01, National Observatory of Sport, Tunis 1003, Tunisia
- Sport and Exercise Science, School of Allied Health, Human Services and Sport, La Trobe University, Melbourne 3086, Australia
| | - Rihab Abid
- High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
| | - Atyh Hadadi
- Department of Sport Sciences, College of Education, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Turki Mohsen Alzahrani
- Department of Sport Sciences, College of Education, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Omar Trabelsi
- High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
- Research Laboratory, Education, Motricity, Sport and Health (EM2S), LR15JS01, High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
| | | | - Liwa Masmoudi
- High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
- Research Laboratory, Education, Motricity, Sport and Health (EM2S), LR15JS01, High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
| | - Ibrahim Ouergui
- High Institute of Sport and Physical Education of Kef, University of Jendouba, El Kef 7100, Tunisia
| | - Kamel Jamoussi
- Laboratory of Biochemistry, CHU Hedi Chaker, University of Sfax, Sfax 3000, Tunisia
| | - Mouna Mnif
- Department of Endocrinology, Hedi Chaker University Hospital of Sfax, Sfax 3038, Tunisia
| | - Hafedh Mejdoub
- Laboratory of Plant Biotechnology, Sfax Faculty of Sciences, BP 1171, University of Sfax, Sfax 3038, Tunisia
| | - Piotr Zmijewski
- Jozef Pilsudski University of Physical Education in Warsaw, 00-809 Warsaw, Poland
| | - Jordan M. Glenn
- Department of Health, Exercise Science Research Center Human Performance and Recreation, University of Arkansas, Fayetteville, AR 72701, USA
| | - Khaled Trabelsi
- High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
- Research Laboratory, Education, Motricity, Sport and Health (EM2S), LR15JS01, High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
| | - Hamdi Chtourou
- High Institute of Sport and Physical Education, University of Sfax, Sfax 3038, Tunisia
- Physical Activity, Sport, and Health, UR18JS01, National Observatory of Sport, Tunis 1003, Tunisia
| |
Collapse
|
23
|
Sheng CY, Son YH, Jang J, Park SJ. In vitro skeletal muscle models for type 2 diabetes. BIOPHYSICS REVIEWS 2022; 3:031306. [PMID: 36124295 PMCID: PMC9478902 DOI: 10.1063/5.0096420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Type 2 diabetes mellitus, a metabolic disorder characterized by abnormally elevated blood sugar, poses a growing social, economic, and medical burden worldwide. The skeletal muscle is the largest metabolic organ responsible for glucose homeostasis in the body, and its inability to properly uptake sugar often precedes type 2 diabetes. Although exercise is known to have preventative and therapeutic effects on type 2 diabetes, the underlying mechanism of these beneficial effects is largely unknown. Animal studies have been conducted to better understand the pathophysiology of type 2 diabetes and the positive effects of exercise on type 2 diabetes. However, the complexity of in vivo systems and the inability of animal models to fully capture human type 2 diabetes genetics and pathophysiology are two major limitations in these animal studies. Fortunately, in vitro models capable of recapitulating human genetics and physiology provide promising avenues to overcome these obstacles. This review summarizes current in vitro type 2 diabetes models with focuses on the skeletal muscle, interorgan crosstalk, and exercise. We discuss diabetes, its pathophysiology, common in vitro type 2 diabetes skeletal muscle models, interorgan crosstalk type 2 diabetes models, exercise benefits on type 2 diabetes, and in vitro type 2 diabetes models with exercise.
Collapse
Affiliation(s)
- Christina Y. Sheng
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Young Hoon Son
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | - Sung-Jin Park
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
24
|
Stocks B, Zierath JR. Post-translational Modifications: The Signals at the Intersection of Exercise, Glucose Uptake, and Insulin Sensitivity. Endocr Rev 2022; 43:654-677. [PMID: 34730177 PMCID: PMC9277643 DOI: 10.1210/endrev/bnab038] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Indexed: 11/19/2022]
Abstract
Diabetes is a global epidemic, of which type 2 diabetes makes up the majority of cases. Nonetheless, for some individuals, type 2 diabetes is eminently preventable and treatable via lifestyle interventions. Glucose uptake into skeletal muscle increases during and in recovery from exercise, with exercise effective at controlling glucose homeostasis in individuals with type 2 diabetes. Furthermore, acute and chronic exercise sensitizes skeletal muscle to insulin. A complex network of signals converge and interact to regulate glucose metabolism and insulin sensitivity in response to exercise. Numerous forms of post-translational modifications (eg, phosphorylation, ubiquitination, acetylation, ribosylation, and more) are regulated by exercise. Here we review the current state of the art of the role of post-translational modifications in transducing exercise-induced signals to modulate glucose uptake and insulin sensitivity within skeletal muscle. Furthermore, we consider emerging evidence for noncanonical signaling in the control of glucose homeostasis and the potential for regulation by exercise. While exercise is clearly an effective intervention to reduce glycemia and improve insulin sensitivity, the insulin- and exercise-sensitive signaling networks orchestrating this biology are not fully clarified. Elucidation of the complex proteome-wide interactions between post-translational modifications and the associated functional implications will identify mechanisms by which exercise regulates glucose homeostasis and insulin sensitivity. In doing so, this knowledge should illuminate novel therapeutic targets to enhance insulin sensitivity for the clinical management of type 2 diabetes.
Collapse
Affiliation(s)
- Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.,Departments of Molecular Medicine and Surgery and Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Frankenberg NT, Mason SA, Wadley GD, Murphy RM. Skeletal muscle cell-specific differences in type 2 diabetes. Cell Mol Life Sci 2022; 79:256. [PMID: 35460430 PMCID: PMC9035013 DOI: 10.1007/s00018-022-04265-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 03/03/2022] [Accepted: 03/20/2022] [Indexed: 11/26/2022]
Abstract
Major stores of glucose are found as glycogen in skeletal muscle and liver. Skeletal muscle is a heterogenous tissue, with cellular metabolic and contractile distinctions dependent on whether the cell (fibre) is slow-twitch (Type I) or fast-twitch (Type II). We hypothesised that proteins important for glycogen metabolism would be differentially abundant between these diverse fibres. We further hypothesised that the cellular location of these proteins would be different in muscle samples between control (CON) and individuals with type 2 diabetes (T2D). We dissected individual muscle fibre segments from vastus lateralis skeletal muscle biopsy samples from CON and T2D and used cell-type-specific approaches to address muscle heterogeneity. We measured glycogen and glycogen-related proteins by immunoblotting techniques. A lower proportion of Type I fibres was found in muscle in T2D compared with CON. AMPK-β2, glycogen branching enzyme (GBE), glycogen debranching enzyme (GDE), and glycogen phosphorylase (GP) were differentially localized between fibre types and in fibres from CON and T2D individuals. A key novel finding was that the majority of glycogen is loosely bound or cytosolic in location in human skeletal muscle. The proportion of this diffusible pool of glycogen was significantly lower in Type I fibres in T2D compared to CON. A hyperinsulinaemic, euglycaemic clamp in people with type 2 diabetes had no effect on the proportion of diffusible glycogen. We identify cell-type as an important consideration when assessing glycogen metabolism in muscle. Our findings demonstrate varying glucose handling abilities in specific muscle fibre types in type 2 diabetes. A model is presented to provide an overview of the cell-specific differences in glycogen metabolism in type 2 diabetes.
Collapse
Affiliation(s)
- Noni T Frankenberg
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, 3086, Australia
| | - Shaun A Mason
- Institute for Physical Activity and Nutrition, Deakin University, Burwood, 3125, Australia
| | - Glenn D Wadley
- Institute for Physical Activity and Nutrition, Deakin University, Burwood, 3125, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, 3086, Australia.
| |
Collapse
|
26
|
Kanaley JA, Colberg SR, Corcoran MH, Malin SK, Rodriguez NR, Crespo CJ, Kirwan JP, Zierath JR. Exercise/Physical Activity in Individuals with Type 2 Diabetes: A Consensus Statement from the American College of Sports Medicine. Med Sci Sports Exerc 2022; 54:353-368. [PMID: 35029593 PMCID: PMC8802999 DOI: 10.1249/mss.0000000000002800] [Citation(s) in RCA: 321] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
ABSTRACT This consensus statement is an update of the 2010 American College of Sports Medicine position stand on exercise and type 2 diabetes. Since then, a substantial amount of research on select topics in exercise in individuals of various ages with type 2 diabetes has been published while diabetes prevalence has continued to expand worldwide. This consensus statement provides a brief summary of the current evidence and extends and updates the prior recommendations. The document has been expanded to include physical activity, a broader, more comprehensive definition of human movement than planned exercise, and reducing sedentary time. Various types of physical activity enhance health and glycemic management in people with type 2 diabetes, including flexibility and balance exercise, and the importance of each recommended type or mode are discussed. In general, the 2018 Physical Activity Guidelines for Americans apply to all individuals with type 2 diabetes, with a few exceptions and modifications. People with type 2 diabetes should engage in physical activity regularly and be encouraged to reduce sedentary time and break up sitting time with frequent activity breaks. Any activities undertaken with acute and chronic health complications related to diabetes may require accommodations to ensure safe and effective participation. Other topics addressed are exercise timing to maximize its glucose-lowering effects and barriers to and inequities in physical activity adoption and maintenance.
Collapse
Affiliation(s)
- Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO
| | - Sheri R Colberg
- Human Movement Sciences Department, Old Dominion University, Norfolk, VA
| | | | - Steven K Malin
- Department of Kinesiology and Health, Rutgers University, New Brunswick, NJ
| | - Nancy R Rodriguez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT
| | - Carlos J Crespo
- Oregon Health and Science University-Portland State University School of Public Health, Portland, OR
| | - John P Kirwan
- Pennington Biomedical Research Center, Baton Rouge, LA
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, SWEDEN
| |
Collapse
|
27
|
Wang CC, Chen HJ, Chan DC, Chiu CY, Liu SH, Lan KC. Low-Dose Acrolein, an Endogenous and Exogenous Toxic Molecule, Inhibits Glucose Transport via an Inhibition of Akt-Regulated GLUT4 Signaling in Skeletal Muscle Cells. Int J Mol Sci 2021; 22:ijms22137228. [PMID: 34281282 PMCID: PMC8268984 DOI: 10.3390/ijms22137228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 01/28/2023] Open
Abstract
Urinary acrolein adduct levels have been reported to be increased in both habitual smokers and type-2 diabetic patients. The impairment of glucose transport in skeletal muscles is a major factor responsible for glucose uptake reduction in type-2 diabetic patients. The effect of acrolein on glucose metabolism in skeletal muscle remains unclear. Here, we investigated whether acrolein affects muscular glucose metabolism in vitro and glucose tolerance in vivo. Exposure of mice to acrolein (2.5 and 5 mg/kg/day) for 4 weeks substantially increased fasting blood glucose and impaired glucose tolerance. The glucose transporter-4 (GLUT4) protein expression was significantly decreased in soleus muscles of acrolein-treated mice. The glucose uptake was significantly decreased in differentiated C2C12 myotubes treated with a non-cytotoxic dose of acrolein (1 μM) for 24 and 72 h. Acrolein (0.5–2 μM) also significantly decreased the GLUT4 expression in myotubes. Acrolein suppressed the phosphorylation of glucose metabolic signals IRS1, Akt, mTOR, p70S6K, and GSK3α/β. Over-expression of constitutive activation of Akt reversed the inhibitory effects of acrolein on GLUT4 protein expression and glucose uptake in myotubes. These results suggest that acrolein at doses relevant to human exposure dysregulates glucose metabolism in skeletal muscle cells and impairs glucose tolerance in mice.
Collapse
Affiliation(s)
- Ching-Chia Wang
- Department of Pediatrics, College of Medicine, National Taiwan University & Hospital, Taipei 100, Taiwan;
| | - Huang-Jen Chen
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Ding-Cheng Chan
- Department of Geriatrics and Gerontology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Chen-Yuan Chiu
- Center of Consultation, Center for Drug Evaluation, Taipei 115, Taiwan;
| | - Shing-Hwa Liu
- Department of Pediatrics, College of Medicine, National Taiwan University & Hospital, Taipei 100, Taiwan;
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
- Correspondence: (S.-H.L.); (K.-C.L.)
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Correspondence: (S.-H.L.); (K.-C.L.)
| |
Collapse
|
28
|
Shur NF, Creedon L, Skirrow S, Atherton PJ, MacDonald IA, Lund J, Greenhaff PL. Age-related changes in muscle architecture and metabolism in humans: The likely contribution of physical inactivity to age-related functional decline. Ageing Res Rev 2021; 68:101344. [PMID: 33872778 PMCID: PMC8140403 DOI: 10.1016/j.arr.2021.101344] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/15/2021] [Accepted: 04/13/2021] [Indexed: 12/21/2022]
Abstract
In the United Kingdom (UK), it is projected that by 2035 people aged >65 years will make up 23 % of the population, with those aged >85 years accounting for 5% of the total population. Ageing is associated with progressive changes in muscle metabolism and a decline in functional capacity, leading to a loss of independence. Muscle metabolic changes associated with ageing have been linked to alterations in muscle architecture and declines in muscle mass and insulin sensitivity. However, the biological features often attributed to muscle ageing are also seen in controlled studies of physical inactivity (e.g. reduced step-count and bed-rest), and it is currently unclear how many of these ageing features are due to ageing per se or sedentarism. This is particularly relevant at a time of home confinements reducing physical activity levels during the Covid-19 pandemic. Current knowledge gaps include the relative contribution that physical inactivity plays in the development of many of the negative features associated with muscle decline in older age. Similarly, data demonstrating positive effects of government recommended physical activity guidelines on muscle health are largely non-existent. It is imperative therefore that research examining interactions between ageing, physical activity and muscle mass and metabolic health is prioritised so that it can inform on the "normal" muscle ageing process and on strategies for improving health span and well-being. This review will focus on important changes in muscle architecture and metabolism that accompany ageing and highlight the likely contribution of physical inactivity to these changes.
Collapse
Affiliation(s)
- N F Shur
- Versus Arthritis Centre for Sport, Exercise and Osteoarthritis, The University of Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, UK; School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - L Creedon
- MRC/Versus Arthritis Centre for Musculoskeletal Ageing Research, UK; School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - S Skirrow
- MRC/Versus Arthritis Centre for Musculoskeletal Ageing Research, UK; School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - P J Atherton
- MRC/Versus Arthritis Centre for Musculoskeletal Ageing Research, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, UK; School of Medicine, University of Nottingham Medical School, Royal Derby Hospital, Derby DE22 3DT, UK
| | - I A MacDonald
- MRC/Versus Arthritis Centre for Musculoskeletal Ageing Research, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, UK; School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK
| | - J Lund
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, UK; School of Medicine, University of Nottingham Medical School, Royal Derby Hospital, Derby DE22 3DT, UK
| | - P L Greenhaff
- MRC/Versus Arthritis Centre for Musculoskeletal Ageing Research, UK; Versus Arthritis Centre for Sport, Exercise and Osteoarthritis, The University of Nottingham, UK; National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, UK; School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, UK.
| |
Collapse
|
29
|
Abstract
As the principal tissue for insulin-stimulated glucose disposal, skeletal muscle is a primary driver of whole-body glycemic control. Skeletal muscle also uniquely responds to muscle contraction or exercise with increased sensitivity to subsequent insulin stimulation. Insulin's dominating control of glucose metabolism is orchestrated by complex and highly regulated signaling cascades that elicit diverse and unique effects on skeletal muscle. We discuss the discoveries that have led to our current understanding of how insulin promotes glucose uptake in muscle. We also touch upon insulin access to muscle, and insulin signaling toward glycogen, lipid, and protein metabolism. We draw from human and rodent studies in vivo, isolated muscle preparations, and muscle cell cultures to home in on the molecular, biophysical, and structural elements mediating these responses. Finally, we offer some perspective on molecular defects that potentially underlie the failure of muscle to take up glucose efficiently during obesity and type 2 diabetes.
Collapse
|
30
|
Jorquera G, Russell J, Monsalves-Álvarez M, Cruz G, Valladares-Ide D, Basualto-Alarcón C, Barrientos G, Estrada M, Llanos P. NLRP3 Inflammasome: Potential Role in Obesity Related Low-Grade Inflammation and Insulin Resistance in Skeletal Muscle. Int J Mol Sci 2021; 22:ijms22063254. [PMID: 33806797 PMCID: PMC8005007 DOI: 10.3390/ijms22063254] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/07/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Among multiple mechanisms, low-grade inflammation is critical for the development of insulin resistance as a feature of type 2 diabetes. The nucleotide-binding oligomerization domain-like receptor family (NOD-like) pyrin domain containing 3 (NLRP3) inflammasome has been linked to the development of insulin resistance in various tissues; however, its role in the development of insulin resistance in the skeletal muscle has not been explored in depth. Currently, there is limited evidence that supports the pathological role of NLRP3 inflammasome activation in glucose handling in the skeletal muscle of obese individuals. Here, we have centered our focus on insulin signaling in skeletal muscle, which is the main site of postprandial glucose disposal in humans. We discuss the current evidence showing that the NLRP3 inflammasome disturbs glucose homeostasis. We also review how NLRP3-associated interleukin and its gasdermin D-mediated efflux could affect insulin-dependent intracellular pathways. Finally, we address pharmacological NLRP3 inhibitors that may have a therapeutical use in obesity-related metabolic alterations.
Collapse
Affiliation(s)
- Gonzalo Jorquera
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.J.); (G.C.)
| | - Javier Russell
- Escuela de Pedagogía en Educación Física, Facultad de Educación, Universidad Autónoma de Chile, Santiago 8900000, Chile;
| | - Matías Monsalves-Álvarez
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (M.M.-Á.); (D.V.-I.)
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (G.J.); (G.C.)
| | - Denisse Valladares-Ide
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2820000, Chile; (M.M.-Á.); (D.V.-I.)
| | - Carla Basualto-Alarcón
- Departamento de Ciencias de la Salud, Universidad de Aysén, Coyhaique 5951537, Chile;
- Departamento de Anatomía y Medicina Legal, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Genaro Barrientos
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (G.B.); (M.E.)
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Manuel Estrada
- Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (G.B.); (M.E.)
| | - Paola Llanos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago 8380544, Chile
- Correspondence: ; Tel.: +56-229-781-727
| |
Collapse
|
31
|
Joseph JS, Anand K, Malindisa ST, Oladipo AO, Fagbohun OF. Exercise, CaMKII, and type 2 diabetes. EXCLI JOURNAL 2021; 20:386-399. [PMID: 33746668 PMCID: PMC7975583 DOI: 10.17179/excli2020-3317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
Individuals who exercise regularly are protected from type 2 diabetes and other metabolic syndromes, in part by enhanced gene transcription and induction of many signaling pathways crucial in correcting impaired metabolic pathways associated with a sedentary lifestyle. Exercise activates Calmodulin-dependent protein kinase (CaMK)II, resulting in increased mitochondrial oxidative capacity and glucose transport. CaMKII regulates many health beneficial cellular functions in individuals who exercise compared with those who do not exercise. The role of exercise in the regulation of carbohydrate, lipid metabolism, and insulin signaling pathways are explained at the onset. Followed by the role of exercise in the regulation of glucose transporter (GLUT)4 expression and mitochondrial biogenesis are explained. Next, the main functions of Calmodulin-dependent protein kinase and the mechanism to activate it are illustrated, finally, an overview of the role of CaMKII in regulating GLUT4 expression, mitochondrial biogenesis, and histone modification are discussed.
Collapse
Affiliation(s)
- Jitcy S. Joseph
- Department of Toxicology and Biochemistry, National Institute for Occupational Health, A division of National Health Laboratory Service, Johannesburg, South Africa
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences and National Health Laboratory Service, University of the Free State, Bloemfontein, South Africa
| | - Sibusiso T. Malindisa
- Department of Life and Consumer Sciences, University of South Africa (UNISA), Florida Park, Johannesburg, South Africa
| | - Adewale O. Oladipo
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering and Technology, University of South Africa, Science Park Florida, Johannesburg, 1710, South Africa
| | - Oladapo F. Fagbohun
- Department of Biomedical Engineering, First Technical University, Ibadan, Oyo State, Nigeria
- Department of Pediatrics, Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
32
|
Laine S, Högel H, Ishizu T, Toivanen J, Yli-Karjanmaa M, Grönroos TJ, Rantala J, Mäkelä R, Hannukainen JC, Kalliokoski KK, Heinonen I. Effects of Different Exercise Training Protocols on Gene Expression of Rac1 and PAK1 in Healthy Rat Fast- and Slow-Type Muscles. Front Physiol 2020; 11:584661. [PMID: 33329033 PMCID: PMC7711069 DOI: 10.3389/fphys.2020.584661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/19/2020] [Indexed: 11/13/2022] Open
Abstract
Purpose Rac1 and its downstream target PAK1 are novel regulators of insulin and exercise-induced glucose uptake in skeletal muscle. However, it is not yet understood how different training intensities affect the expression of these proteins. Therefore, we studied the effects of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on Rac1 and PAK1 expression in fast-type (gastrocnemius, GC) and slow-type (soleus, SOL) muscles in rats after HIIT and MICT swimming exercises. Methods The mRNA expression was determined using qPCR and protein expression levels with reverse-phase protein microarray (RPPA). Results HIIT significantly decreased Rac1 mRNA expression in GC compared to MICT (p = 0.003) and to the control group (CON) (p = 0.001). At the protein level Rac1 was increased in GC in both training groups, but only the difference between HIIT and CON was significant (p = 0.02). HIIT caused significant decrease of PAK1 mRNA expression in GC compared to MICT (p = 0.007) and to CON (p = 0.001). At the protein level, HIIT increased PAK1 expression in GC compared to MICT and CON (by ∼17%), but the difference was not statistically significant (p = 0.3, p = 0.2, respectively). There were no significant differences in the Rac1 or PAK1 expression in SOL between the groups. Conclusion Our results indicate that HIIT, but not MICT, decreases Rac1 and PAK1 mRNA expression and increases the protein expression of especially Rac1 but only in fast-type muscle. These exercise training findings may reveal new therapeutic targets to treat patients with metabolic diseases.
Collapse
Affiliation(s)
- Saara Laine
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Heidi Högel
- Turku Centre for Biotechnology, University of Turku, Åbo Akademi University, Turku, Finland.,Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Tamiko Ishizu
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland.,Institute of Biomedicine, University of Turku, Turku, Finland.,TuDMM Doctoral Programmes, University of Turku, Turku, Finland
| | - Jussi Toivanen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Minna Yli-Karjanmaa
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Tove J Grönroos
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland.,MediCity Research Laboratory, University of Turku, Turku, Finland
| | | | | | - Jarna C Hannukainen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
| | - Kari K Kalliokoski
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
| | - Ilkka Heinonen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland.,Rydberg Laboratory of Applied Sciences, University of Halmstad, Halmstad, Sweden
| |
Collapse
|
33
|
Mok J, Park TS, Kim S, Kim D, Choi CS, Park J. Prokineticin receptor 1 ameliorates insulin resistance in skeletal muscle. FASEB J 2020; 35:e21179. [PMID: 33184929 DOI: 10.1096/fj.202001641r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/17/2020] [Accepted: 10/26/2020] [Indexed: 01/05/2023]
Abstract
Type 2 diabetes mellitus may result from insulin resistance in skeletal muscle. Prokineticin receptor 1 (Prokr1) improves metabolic phenotype in adipose tissue and the cardiovascular system; however, its effects on skeletal muscle have not been investigated. We investigated the Prokr1 signaling pathways and its metabolic function in murine myoblast, satellite cells, and their differentiated myotubes. We measured the expression levels of Prokr1 in the skeletal muscle of mice as well as human skeletal muscle cell-derived myotubes. Prokineticin 2 (PROK2), a ligand of PROKR1, induced calcium mobilization in a dose-dependent manner and altered the mRNA levels of 578 genes in PROKR1-overexpressed HEK293T cells. Functional enrichment of differentially expressed genes revealed that PROKR1 activated Gq-mediated PI3K/AKT and MAPK/ERK signaling pathways in skeletal muscle cells. Prokr1 significantly activated the PI3K/AKT signaling pathway in myotubes derived from C2C12 and satellite cells, regardless of the presence or absence of insulin. Prokr1 also promoted the translocation of glucose transporter 4 (GLUT4) into the plasma membrane. In palmitate-induced insulin-resistant myotubes, Prokr1 enhanced insulin-stimulated AKT phosphorylation, GLUT4 translocation, and glucose uptake. mRNA and protein levels of Prokr1 were significantly decreased in skeletal muscle and white adipose tissue of diet-induced obese mice, and the amount of PROKR1 protein was significantly decreased in human skeletal muscle cell-derived myotubes under insulin resistance conditions. Taken together, these results demonstrate that Prokr1 plays an important role in insulin sensitivity and is a potential therapeutic target to ameliorate insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Jongsoo Mok
- Department of International Agricultural Technology, Graduate School of International Agricultural Technology, Seoul National University, Seoul, Republic of Korea
| | - Tae Sub Park
- Department of International Agricultural Technology, Graduate School of International Agricultural Technology, Seoul National University, Seoul, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Sunhong Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongwon-gun, Republic of Korea.,Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Daehoon Kim
- Pharmacology Research Division, New Drug Discovery Center, Hyundai Pharm Co, Cheonan-si, Republic of Korea
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Seongnam-si, Republic of Korea.,Endocrinology, Internal Medicine, Gachon University Gil Medical Center, Seongnam-si, Republic of Korea
| | - Joonghoon Park
- Department of International Agricultural Technology, Graduate School of International Agricultural Technology, Seoul National University, Seoul, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
34
|
Batista TM, Jayavelu AK, Wewer Albrechtsen NJ, Iovino S, Lebastchi J, Pan H, Dreyfuss JM, Krook A, Zierath JR, Mann M, Kahn CR. A Cell-Autonomous Signature of Dysregulated Protein Phosphorylation Underlies Muscle Insulin Resistance in Type 2 Diabetes. Cell Metab 2020; 32:844-859.e5. [PMID: 32888406 PMCID: PMC7875546 DOI: 10.1016/j.cmet.2020.08.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/26/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Skeletal muscle insulin resistance is the earliest defect in type 2 diabetes (T2D), preceding and predicting disease development. To what extent this reflects a primary defect or is secondary to tissue cross talk due to changes in hormones or circulating metabolites is unknown. To address this question, we have developed an in vitro disease-in-a-dish model using iPS cells from T2D patients differentiated into myoblasts (iMyos). We find that T2D iMyos in culture exhibit multiple defects mirroring human disease, including an altered insulin signaling, decreased insulin-stimulated glucose uptake, and reduced mitochondrial oxidation. More strikingly, global phosphoproteomic analysis reveals a multidimensional network of signaling defects in T2D iMyos going beyond the canonical insulin-signaling cascade, including proteins involved in regulation of Rho GTPases, mRNA splicing and/or processing, vesicular trafficking, gene transcription, and chromatin remodeling. These cell-autonomous defects and the dysregulated network of protein phosphorylation reveal a new dimension in the cellular mechanisms underlying the fundamental defects in T2D.
Collapse
Affiliation(s)
- Thiago M Batista
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ashok Kumar Jayavelu
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Nicolai J Wewer Albrechtsen
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen 2100, Denmark
| | - Salvatore Iovino
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jasmin Lebastchi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Section of Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 76, Sweden
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
35
|
Larsen MR, Steenberg DE, Birk JB, Sjøberg KA, Kiens B, Richter EA, Wojtaszewski JFP. The insulin‐sensitizing effect of a single exercise bout is similar in type I and type II human muscle fibres. J Physiol 2020; 598:5687-5699. [DOI: 10.1113/jp280475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/02/2020] [Indexed: 11/08/2022] Open
Affiliation(s)
- Magnus R. Larsen
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports Faculty of Science University of Copenhagen Copenhagen Denmark
| | - Dorte E. Steenberg
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports Faculty of Science University of Copenhagen Copenhagen Denmark
| | - Jesper B. Birk
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports Faculty of Science University of Copenhagen Copenhagen Denmark
| | - Kim A. Sjøberg
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports Faculty of Science University of Copenhagen Copenhagen Denmark
| | - Bente Kiens
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports Faculty of Science University of Copenhagen Copenhagen Denmark
| | - Erik A. Richter
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports Faculty of Science University of Copenhagen Copenhagen Denmark
| | - Jørgen F. P. Wojtaszewski
- Section of Molecular Physiology Department of Nutrition, Exercise and Sports Faculty of Science University of Copenhagen Copenhagen Denmark
| |
Collapse
|
36
|
Are Cape Peninsula baboons raiding their way to obesity and type II diabetes? - a comparative study. Comp Biochem Physiol A Mol Integr Physiol 2020; 250:110794. [PMID: 32827764 DOI: 10.1016/j.cbpa.2020.110794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/16/2020] [Accepted: 08/16/2020] [Indexed: 01/20/2023]
Abstract
Researchers, managers and conservationists in the Cape Peninsula, South Africa, have reported cases of individual baboons (Papio ursinus) appearing overweight, lethargic and having poor teeth. Despite an intensive baboon management programme, there are certain individual baboons and troops that continue to raid human food sources. These food sources often are high in processed carbohydrates and saturated fats. As this diet is highly associated with obesity, insulin resistance and type II diabetes, the present study aimed to establish if these baboons may be at risk of developing insulin resistance. Post mortem muscle samples from 17 Cape Peninsula and 7 control adult male baboons were rapidly frozen in liquid nitrogen and analysed for insulin receptor substrate-1 (IRS-1), glucose transporter 4 (GLUT4), oxidative and glycolytic markers of metabolism (citrate synthase, 3-hydroxyacyl-CoA-dehydrogenase, lactate dehydrogenase and creatine kinase activities), and muscle fibre morphology. The sampled Peninsula baboons were heavier (33 ± 2 vs. 29 ± 2 kg, P < 0.05) and had a higher frequency of poor teeth compared to control baboons. Muscle fibre type, fibre size, GLUT4 content, oxidative and glycolytic metabolism were not different between the two groups. However, IRS-1 content, a marker of insulin sensitivity, was significantly lower (by 43%, P < 0.001) in the Peninsula baboons compared to the controls. This study provides the first indirect evidence that some Peninsula baboons with a history of raiding human food sources, may be at risk of developing insulin resistance in the wild, with long term implications for population health.
Collapse
|
37
|
Vesentini G, Barbosa AMP, Floriano JF, Felisbino SL, Costa SMB, Piculo F, Marini G, Nunes SK, Reyes DRA, Marcondes JPC, Hallur RLS, Rozza AL, Magalhães CG, Costa R, Abbade JF, Corrente JE, Calderon IMP, Matheus SMM, Rudge MVC. Deleterious effects of gestational diabetes mellitus on the characteristics of the rectus abdominis muscle associated with pregnancy-specific urinary incontinence. Diabetes Res Clin Pract 2020; 166:108315. [PMID: 32679058 DOI: 10.1016/j.diabres.2020.108315] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/05/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
AIMS To evaluate the effects of gestational diabetes mellitus (GDM) on the structural characteristics of the rectus abdominis muscle (RAM) and its indirect effects on pregnancy-specific urinary incontinence (PSUI). METHODS A total of 92 pregnant women were divided into four groups, according to their clinical conditions: non-GDM continent, non-GDM associated PSUI, GDM continent and GDM associated PSUI. The muscle morphometry (histochemistry and immunohistochemistry) for the fiber types and collagen fiber distribution, the ultrastructural analysis (transmission electron microscopy), the protein expression of fiber types and calcium signaling (Western blotting), and the content of types I and III collagen fiber (ELISA) in RAM collected at delivery were assessed. RESULTS The GDM groups presented a significantly increased number of slow fibers and slow-twitch oxidative fiber expression; decreased fiber area, number of fast fibers, and area of collagen; an increase in central nuclei; ultrastructural alterations with focal lesion areas such as myeloid structures, sarcomere disorganization, and mitochondrial alteration. The PSUI groups presented a considerable decrease in types I and III collagen contents and the localization of collagen fiber. CONCLUSIONS Our data reveal that GDM causes morphological, biochemical and physiological changes in the RAM, and this might predispose women to PSUI.
Collapse
Affiliation(s)
- Giovana Vesentini
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Angélica M P Barbosa
- São Paulo State University (UNESP), School of Philosophy and Sciences, Department of Physical Therapy and Occupational Therapy, Marilia, São Paulo State, Brazil
| | - Juliana F Floriano
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Sérgio L Felisbino
- São Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu, São Paulo State, Brazil
| | - Sarah M B Costa
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Fernanda Piculo
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Gabriela Marini
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil; Universidade Sagrado Coração, Department of Health Sciences, Bauru, São Paulo, Brazil
| | - Sthefanie K Nunes
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - David R A Reyes
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - João P C Marcondes
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Raghavendra L S Hallur
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Ariane L Rozza
- São Paulo State University (UNESP), Institute of Biosciences, Department of Morphology, Botucatu, São Paulo State, Brazil
| | - Cláudia G Magalhães
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Roberto Costa
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Joelcio F Abbade
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - José E Corrente
- São Paulo State University (UNESP), Institute of Biosciences, Biostatistics Department, Botucatu, São Paulo, Brazil
| | - Iracema M P Calderon
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil
| | - Selma M M Matheus
- São Paulo State University (UNESP), Institute of Biosciences, Department of Anatomy, Botucatu, São Paulo State, Brazil
| | - Marilza V C Rudge
- Perinatal Diabetes Research Center, University Hospital, Botucatu Medical School, Univ Estadual Paulista_UNESP, São Paulo State, Brazil; São Paulo State University (UNESP), Botucatu Medical School, Department of Gynecology and Obstetrics, Botucatu, Sao Paulo, Brazil.
| |
Collapse
|
38
|
Schiffer TA, Lundberg JO, Weitzberg E, Carlström M. Modulation of mitochondria and NADPH oxidase function by the nitrate-nitrite-NO pathway in metabolic disease with focus on type 2 diabetes. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165811. [PMID: 32339643 DOI: 10.1016/j.bbadis.2020.165811] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/16/2020] [Accepted: 04/18/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria play fundamental role in maintaining cellular metabolic homeostasis, and metabolic disorders including type 2 diabetes (T2D) have been associated with mitochondrial dysfunction. Pathophysiological mechanisms are coupled to increased production of reactive oxygen species and oxidative stress, together with reduced bioactivity/signaling of nitric oxide (NO). Novel strategies restoring these abnormalities may have therapeutic potential in order to prevent or even treat T2D and associated cardiovascular and renal co-morbidities. A diet rich in green leafy vegetables, which contains high concentrations of inorganic nitrate, has been shown to reduce the risk of T2D. To this regard research has shown that in addition to the classical NO synthase (NOS) dependent pathway, nitrate from our diet can work as an alternative precursor for NO and other bioactive nitrogen oxide species via serial reductions of nitrate (i.e. nitrate-nitrite-NO pathway). This non-conventional pathway may act as an efficient back-up system during various pathological conditions when the endogenous NOS system is compromised (e.g. acidemia, hypoxia, ischemia, aging, oxidative stress). A number of experimental studies have demonstrated protective effects of nitrate supplementation in models of obesity, metabolic syndrome and T2D. Recently, attention has been directed towards the effects of nitrate/nitrite on mitochondrial functions including beiging/browning of white adipose tissue, PGC-1α and SIRT3 dependent AMPK activation, GLUT4 translocation and mitochondrial fusion-dependent improvements in glucose homeostasis, as well as dampening of NADPH oxidase activity. In this review, we examine recent research related to the effects of bioactive nitrogen oxide species on mitochondrial function with emphasis on T2D.
Collapse
Affiliation(s)
- Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
39
|
Diabetic Cardiomyopathy and Ischemic Heart Disease: Prevention and Therapy by Exercise and Conditioning. Int J Mol Sci 2020; 21:ijms21082896. [PMID: 32326182 PMCID: PMC7215312 DOI: 10.3390/ijms21082896] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/14/2020] [Accepted: 04/18/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome, diabetes, and ischemic heart disease are among the leading causes of death and disability in Western countries. Diabetic cardiomyopathy is responsible for the most severe signs and symptoms. An important strategy for reducing the incidence of cardiovascular disease is regular exercise. Remote ischemic conditioning has some similarity with exercise and can be induced by short periods of ischemia and reperfusion of a limb, and it can be performed in people who cannot exercise. There is abundant evidence that exercise is beneficial in diabetes and ischemic heart disease, but there is a need to elucidate the specific cardiovascular effects of emerging and unconventional forms of exercise in people with diabetes. In addition, remote ischemic conditioning may be considered among the options to induce beneficial effects in these patients. The characteristics and interactions of diabetes and ischemic heart disease, and the known effects of exercise and remote ischemic conditioning in the presence of metabolic syndrome and diabetes, are analyzed in this brief review.
Collapse
|
40
|
Abstract
AbstractThis study investigated metabolic, endocrine, appetite and mood responses to a maximal eating occasion in fourteen men (mean: age 28 (sd5) years, body mass 77·2 (sd6·6) kg and BMI 24·2 (sd2·2) kg/m2) who completed two trials in a randomised crossover design. On each occasion, participants ate a homogenous mixed-macronutrient meal (pizza). On one occasion, they ate until ‘comfortably full’ (ad libitum) and on the other, until they ‘could not eat another bite’ (maximal). Mean energy intake was double in the maximal (13 024 (95 % CI 10 964, 15 084) kJ; 3113 (95 % CI 2620, 3605) kcal) compared with thead libitumtrial (6627 (95 % CI 5708, 7547) kJ; 1584 (95 % CI 1364, 1804) kcal). Serum insulin incremental AUC (iAUC) increased approximately 1·5-fold in the maximal compared withad libitumtrial (mean:ad libitum43·8 (95 % CI 28·3, 59·3) nmol/l × 240 min and maximal 67·7 (95 % CI 47·0, 88·5) nmol/l × 240 min,P< 0·01), but glucose iAUC did not differ between trials (ad libitum94·3 (95 % CI 30·3, 158·2) mmol/l × 240 min and maximal 126·5 (95 % CI 76·9, 176·0) mmol/l × 240 min,P= 0·19). TAG iAUC was approximately 1·5-fold greater in the maximalv.ad libitumtrial (ad libitum98·6 (95 % CI 69·9, 127·2) mmol/l × 240 min and maximal 146·4 (95 % CI 88·6, 204·1) mmol/l × 240 min,P< 0·01). Total glucagon-like peptide-1, glucose-dependent insulinotropic peptide and peptide tyrosine–tyrosine iAUC were greater in the maximal compared withad libitumtrial (P< 0·05). Total ghrelin concentrations decreased to a similar extent, but AUC was slightly lower in the maximalv.ad libitumtrial (P= 0·02). There were marked differences on appetite and mood between trials, most notably maximal eating caused a prolonged increase in lethargy. Healthy men have the capacity to eat twice the energy content required to achieve comfortable fullness at a single meal. Postprandial glycaemia is well regulated following initial overeating, with elevated postprandial insulinaemia probably contributing.
Collapse
|
41
|
Saha S. Association between the membrane transporter proteins and type 2 diabetes mellitus. Expert Rev Clin Pharmacol 2020; 13:287-297. [PMID: 32066279 DOI: 10.1080/17512433.2020.1729125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: The prevalence rate of diabetes is increasing day by day and the current scenario of the available agents for its treatment has given rise to stimulation in the search for new therapeutic targets and agents. Therefore the present review will examine the role of membrane composition in the pathophysiology of Type 2 Diabetes and the possible therapeutic approaches for this.Areas covered: Glucose transporter proteins (GLUTs) are integral membrane proteins which are responsible for facilitated glucose transport over the plasma membrane into cells. Thus, this chapter is an attempt to interpret the co-relation between membrane transporter proteins and lipid molecules of cell membrane and their implications in type 2 diabetes mellitus. The relationship between the composition controlled flexibility of the membrane in the insertion of GLUTs into cell membrane as well as its fusion with the membrane is the focus of this chapter.Expert opinion: There is increasing data on the central role of phospholipid composition toward T2DM. Plasma membrane lipid composition plays a key role in maintaining the machinery for insulin-independent GLUT insertion into the membrane as well as insulin-dependent GLUT4 containing vesicles. As a therapeutic option, the designing of new chemical entities should be aimed to decrease saturated fatty acids of lipid bilayer phospholipids to target type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Zoology, University School of Sciences, Gujarat University, Ahmedabad, India
| |
Collapse
|
42
|
Minami S, Yokota N, Kawahara H. BAG6 contributes to glucose uptake by supporting the cell surface translocation of the glucose transporter GLUT4. Biol Open 2020; 9:bio.047324. [PMID: 31911483 PMCID: PMC6994957 DOI: 10.1242/bio.047324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Defective translocation of glucose transporter 4 (GLUT4) to the cell surface is a key feature of insulin resistance in type 2 diabetes. Therefore, elucidating the mechanism of GLUT4 translocation is of primary importance. The mammalian Bag6/Bat3 gene has been suggested to be linked with potential obesity- and diabetes-associated loci, while its function in the control of glucose incorporation into the cytoplasm has not been investigated. In this study, we established a series of cell lines that stably expressed GLUT4 with three tandem repeats of the antigenic peptide inserted into its 1st extracellular loop. With these cell lines, we found that the depletion of endogenous BAG6 downregulated the cell surface expression of GLUT4, concomitant with the reduced incorporation of a glucose analog into the cells. Defective intracellular translocation of GLUT4 in BAG6-depleted cells is similar to the case observed for the depletion of Rab8a, an essential regulator of insulin-stimulated GLUT4 translocation. In addition, we observed that the assembly of syntaxin 6 into the endoplasmic reticulum membrane was slightly disturbed under BAG6 depletion. Given that Rab8a and syntaxin 6 are critical for GLUT4 translocation, we suggest that BAG6 may play multiple roles in the trafficking of glucose transporters to the cell surface. This article has an associated First Person interview with the first author of the paper. Summary: BAG6 is critical for the insulin-stimulated translocation of GLUT4 from its peri-nuclear storage compartments to the cell surface.
Collapse
Affiliation(s)
- Setsuya Minami
- Laboratory of Cell Biology and Biochemistry, Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Naoto Yokota
- Laboratory of Cell Biology and Biochemistry, Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Hiroyuki Kawahara
- Laboratory of Cell Biology and Biochemistry, Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
43
|
Abstract
In insulin resistance, alterations occur in the signalling pathways that modulate glucose uptake into cells, especially skeletal muscle cells, resulting in impaired glucose homeostasis. Glucose uptake into cells is controlled by a number of pathways, some of which are insulin-dependent. During exercise glucose uptake can occur independently of insulin regulation, and hence research into the effects of exercise on insulin resistance must be clearly defined to reflect whether glucose uptake has been enhanced as a result of the utilisation of these insulin-independent pathways, or whether exercise directly affects insulin resistance in cells. Research into the benefits of exercise for insulin resistance is also problematic in the need to clarify whether it is the exercise itself, or the visceral fat/weight loss that has resulted from the exercise, that has led to improved insulin sensitivity. The research presents a promising picture for the benefits of exercise in insulin resistance.
Collapse
Affiliation(s)
- Stephney Whillier
- Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
44
|
Computed tomography-based psoas skeletal muscle area and radiodensity are poor sentinels for whole L3 skeletal muscle values. Clin Nutr 2019; 39:2227-2232. [PMID: 31668722 PMCID: PMC7359407 DOI: 10.1016/j.clnu.2019.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 02/07/2023]
Abstract
Background and aims Computed tomography (CT)-based measurement of skeletal muscle cross-sectional area (CSA) and Hounsfield unit (HU) radiodensity are used to assess the presence of sarcopenia and myosteatosis, respectively. The validated CT-based technique involves analysis of skeletal muscle at the third lumbar vertebral (L3) level. Recently there has been increasing interest in the use of psoas muscle alone as a sentinel. However, this technique has not been extensively investigated or compared with the previous validated standard approach. Methods Portovenous phase CT images at the L3 level were identified retrospectively from a single institution in 150 patients who had non-emergency scans and were analysed by a single assessor using SliceOmatic software v5.0 (TomoVision, Canada). Manual segmentation based upon validated HU thresholds for skeletal muscle density was performed for all skeletal muscle, as well as the individual muscle groups. The muscle CSA and mean radiodensity of each group were compared against the whole L3 slice values. Results When compared with whole L3 slice CSA, anterior abdominal wall CSA had the strongest correlation (r = 0.9315, p < 0.0001) followed by paravertebral (r = 0.8948, p < 0.0001), then psoas muscle (r = 0.7041, p < 0.0001). The mean ± SD density of the psoas muscle (42 ± 8.4 HU) was significantly higher than the whole slice radiodensity (32.3 ± 9.5 HU, p < 0.0001), with paravertebral radiodensity being a more accurate estimation (34.5 ± 10.8 HU). There was a significant difference in the prevalence of myosteatosis when the density measured from the psoas was compared with that of the whole L3 skeletal muscle (27.7% vs. 66.0%, p < 0.0001). Conclusion Whole L3 slice CSA correlated positively with psoas muscle CSA but was subject to wide variability in results. Psoas muscle radiodensity was significantly greater than whole L3 slice density and resulted in underestimation of the prevalence of myosteatosis. Given the lack of equivalence from individual muscle groups, we recommend that further work be undertaken to investigate which muscle group, or indeed whether the gold standard of whole L3 skeletal muscle, provides the best correlation with clinical outcomes.
Collapse
|
45
|
Regulation of Skeletal Muscle Glucose Transport and Glucose Metabolism by Exercise Training. Nutrients 2019; 11:nu11102432. [PMID: 31614762 PMCID: PMC6835691 DOI: 10.3390/nu11102432] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 09/30/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
Aerobic exercise training and resistance exercise training are both well-known for their ability to improve human health; especially in individuals with type 2 diabetes. However, there are critical differences between these two main forms of exercise training and the adaptations that they induce in the body that may account for their beneficial effects. This article reviews the literature and highlights key gaps in our current understanding of the effects of aerobic and resistance exercise training on the regulation of systemic glucose homeostasis, skeletal muscle glucose transport and skeletal muscle glucose metabolism.
Collapse
|
46
|
Kowalsky RJ, Jakicic JM, Hergenroeder A, Rogers RJ, Gibbs BB. Acute cardiometabolic effects of interrupting sitting with resistance exercise breaks. Appl Physiol Nutr Metab 2019; 44:1025-1032. [DOI: 10.1139/apnm-2018-0633] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Interrupting prolonged sitting with light activity breaks, such as short walks, improves cardiometabolic outcomes, yet less is known about the impact of resistance exercise breaks. This study examined the effects of hourly, guidelines-based simple resistance exercise breaks on acute cardiometabolic health outcomes over a simulated work period. Fourteen adults (age: 53.4 ± 9.5 years, body mass index: 30.9 ± 4.8 kg/m2) completed 2 randomized 4-h conditions: prolonged sitting (SIT) and hourly resistance exercise breaks (REX). Glucose, triglycerides, blood pressure, and heart rate were measured at baseline and then hourly. Pulse wave velocity (PWV) was measured before and after each condition. Linear mixed models evaluated overall condition effects and differences at each hour. Cohen’s d estimated magnitude of effects. Four-hour glucose area under the curve (AUC) did not differ by condition (REX vs. SIT: β = –0.35 mmol/L, p = 0.278, d = 0.51). However, an attenuation of postprandial glucose at 1 h (β = –0.69 mg/dL, p = 0.004, d = 1.02) in REX compared with SIT was observed. Triglyceride AUC, mean blood pressure, and PWV did not differ significantly between REX and SIT overall or any time point (all p > 0.05). Heart rate was higher across the experimental period in REX versus SIT (β = 3.3 bpm, p < 0.001, d = 0.35) and individual time points (β ≥ 3.2 bpm, p ≤ 0.044, d ≥ 0.34). Resistance exercise breaks can potentially improve 1-h postprandial glucose, but may not acutely benefit other cardiometabolic outcomes. Future studies employing guidelines-based resistance exercises to interrupt prolonged sitting with a larger sample and longer follow-up are warranted.
Collapse
Affiliation(s)
- Robert J. Kowalsky
- Department of Health & Kinesiology, Texas A&M University Kingsville, Kingsville, TX 78363, USA
- Department of Health & Physical Activity, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - John M. Jakicic
- Department of Health & Physical Activity, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Healthy Lifestyle Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Andrea Hergenroeder
- Department of Physical Therapy, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Renee J. Rogers
- Department of Health & Physical Activity, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Healthy Lifestyle Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Bethany Barone Gibbs
- Department of Health & Physical Activity, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
47
|
Chandirasegaran G, Elanchezhiyan C, Ghosh K. Modulatory Effects of Berberine Chloride on Lipid Profile, Oxidant Status and Insulin Signaling Molecules in Streptozotocin Induced Diabetic Rats. Indian J Clin Biochem 2019; 34:254-262. [PMID: 31391714 PMCID: PMC6660533 DOI: 10.1007/s12291-018-0754-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 04/24/2018] [Indexed: 12/31/2022]
Abstract
The goal of the present study was to evaluate the effect of Berberine chloride (BC) on lipid profile, oxidant status and insulin signaling molecules in Streptozotocin (STZ) induced diabetic rat model. Diabetes was induced in rats by a single dose of intraperitoneal administration of STZ (40 mg/kg b.w). Diabetic rats were treated with BC (50 mg/kg b.w) and glibenclamide (6 mg/kg b.w) for 45 days. BC treated diabetic rats showed significant (p <0.05) decrease in the levels of TC, TG, phospholipids, LDL, VLDL and lipid peroxidation markers such as LOOH and TBARS. An increase in enzymatic antioxidant (SOD, CAT and GPx), non-enzymatic antioxidant (GSH, vitamin C and E) and insulin signaling molecules expression, like Insulin receptor substrate-1 (IRS-1), Protein kinase B (PKB or Akt) and glucose transporter-4 (GLUT-4) were found to be significantly raised in BC treated STZ induced diabetic rats. Thus, the results of the current study demonstrated that BC significantly reversed the abnormal levels of lipids, oxidant status and insulin signaling molecules in the diabetic rat model, which may be contributed to its anti-diabetic and antioxidant activities.
Collapse
Affiliation(s)
| | | | - Kavisa Ghosh
- Unit of Toxicology, Department of Zoology, School of Life Sciences, Bharathiar University, Coimbatore, Tamil Nadu 641 046 India
| |
Collapse
|
48
|
Abstract
A pivotal metabolic function of insulin is the stimulation of glucose uptake into muscle and adipose tissues. The discovery of the insulin-responsive glucose transporter type 4 (GLUT4) protein in 1988 inspired its molecular cloning in the following year. It also spurred numerous cellular mechanistic studies laying the foundations for how insulin regulates glucose uptake by muscle and fat cells. Here, we reflect on the importance of the GLUT4 discovery and chronicle additional key findings made in the past 30 years. That exocytosis of a multispanning membrane protein regulates cellular glucose transport illuminated a novel adaptation of the secretory pathway, which is to transiently modulate the protein composition of the cellular plasma membrane. GLUT4 controls glucose transport into fat and muscle tissues in response to insulin and also into muscle during exercise. Thus, investigation of regulated GLUT4 trafficking provides a major means by which to map the essential signaling components that transmit the effects of insulin and exercise. Manipulation of the expression of GLUT4 or GLUT4-regulating molecules in mice has revealed the impact of glucose uptake on whole-body metabolism. Remaining gaps in our understanding of GLUT4 function and regulation are highlighted here, along with opportunities for future discoveries and for the development of therapeutic approaches to manage metabolic disease.
Collapse
Affiliation(s)
- Amira Klip
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Timothy E McGraw
- Department of Biochemistry, Weill Medical College of Cornell University, New York, New York 10065
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia
| |
Collapse
|
49
|
Taderera T, Chagonda LS, Gomo E, Katerere D, Shai LJ. Annona stenophylla aqueous extract stimulate glucose uptake in established C2Cl2 muscle cell lines. Afr Health Sci 2019; 19:2219-2229. [PMID: 31656507 PMCID: PMC6794551 DOI: 10.4314/ahs.v19i2.47] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Annona stenophylla is a folk medicine popularly used in Zimbabwe for the treatment of many ailments. This study was carried out to determine some of the possible anti diabetic mechanisms of its action using in vitro cell culturing methods. METHODS A. stenophylla's effects on glucose uptake were tested using muscle cells (C2Cl2). Expression of glucose 4 transporters was determined by treating cell lines with plant extract. Total RNA was isolated and using RT-PCR, GLUT 4 expression levels were quantified. Translocation of GLUT 4 was assessed using FITC fluorescence measured by flow cytometry. RESULTS Treatment of cells with plant extract significantly increased glucose uptake in a concentration dependent manner, with the highest concentration (250 µg/ml) giving 28% increased uptake compared to the negative control. The increase in glucose uptake (2.5 times more than control) was coupled to increase in GLUT 4 mRNA and subsequently GLUT 4 translocation. Wortmannin expunged the A. stenophylla induced increase in GLUT 4 mRNA and glucose uptake. CONCLUSION The results suggest that A. stenophylla aqueous extract increases glucose uptake partly through increasing the GLUT 4 mRNA and translocation potentially acting via the PI-3-K pathway. This study confirms the ethnopharmacological uses of A. stenophylla indicating potential for anti-diabetic products formulation.
Collapse
|
50
|
Wang Q, Hu J, Liu Y, Li J, Liu B, Li M, Lou S. Aerobic Exercise Improves Synaptic-Related Proteins of Diabetic Rats by Inhibiting FOXO1/NF-κB/NLRP3 Inflammatory Signaling Pathway and Ameliorating PI3K/Akt Insulin Signaling Pathway. J Mol Neurosci 2019; 69:28-38. [DOI: 10.1007/s12031-019-01302-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/14/2019] [Indexed: 01/08/2023]
|