1
|
Bubak MP, Davidyan A, O'Reilly CL, Mondal SA, Keast J, Doidge SM, Borowik AK, Taylor ME, Volovičeva E, Kinter MT, Britton SL, Koch LG, Stout MB, Lewis TL, Miller BF. Metformin treatment results in distinctive skeletal muscle mitochondrial remodeling in rats with different intrinsic aerobic capacities. Aging Cell 2024; 23:e14235. [PMID: 38923664 PMCID: PMC11488331 DOI: 10.1111/acel.14235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/07/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
The rationale for the use of metformin as a treatment to slow aging was largely based on data collected from metabolically unhealthy individuals. For healthspan extension metformin will also be used in periods of good health. To understand the potential context specificity of metformin treatment on skeletal muscle, we used a rat model (high-capacity runner/low-capacity runner [HCR/LCR]) with a divide in intrinsic aerobic capacity. Outcomes of metformin treatment differed based on baseline intrinsic mitochondrial function, oxidative capacity of the muscle (gastroc vs soleus), and the mitochondrial population (intermyofibrillar vs. subsarcolemmal). Metformin caused lower ADP-stimulated respiration in LCRs, with less of a change in HCRs. However, a washout of metformin resulted in an unexpected doubling of respiratory capacity in HCRs. These improvements in respiratory capacity were accompanied by mitochondrial remodeling that included increases in protein synthesis and changes in morphology. Our findings raise questions about whether the positive findings of metformin treatment are broadly applicable.
Collapse
Affiliation(s)
- Matthew P. Bubak
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Arik Davidyan
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Department of Biological SciencesCalifornia State University SacramentoSacramentoCaliforniaUSA
| | - Colleen L. O'Reilly
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Samim A. Mondal
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Jordan Keast
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Stephen M. Doidge
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Agnieszka K. Borowik
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Michael E. Taylor
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Evelina Volovičeva
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Michael T. Kinter
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Steven L. Britton
- Department of AnesthesiologyUniversity of MichiganAnn ArborMichiganUSA
- Department of Molecular & Integrative PhysiologyUniversity of MichiganAnn ArborMichiganUSA
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, College of Medicine and Life SciencesThe University of ToledoToledoOhioUSA
| | - Michael B. Stout
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Tommy L. Lewis
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Benjamin F. Miller
- Aging and Metabolism Research ProgramThe Oklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- The Oklahoma VA Medical CenterOklahoma CityOklahomaUSA
| |
Collapse
|
2
|
Chantarasakha K, Yangchum A, Isaka M, Tepaamorndech S. Fungal Depsidones Stimulate AKT-Dependent Glucose Uptake in 3T3-L1 Adipocytes. JOURNAL OF NATURAL PRODUCTS 2024; 87:1673-1681. [PMID: 38597733 PMCID: PMC11287747 DOI: 10.1021/acs.jnatprod.3c01134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/11/2024]
Abstract
Enhanced glucose uptake in insulin-sensitive tissues is one of the therapeutic strategies to ameliorate hyperglycemia and maintain glucose homeostasis in type 2 diabetes. This study disclosed the role of fungal depsidones in glucose uptake and the underlying mechanism in 3T3-L1 adipocytes. Depsidones, including nidulin, nornidulin, and unguinol, isolated from Aspergillus unguis, stimulate glucose uptake in adipocytes. Compared to the others, nidulin exhibited an upward trend in glucose uptake. The effect of nidulin was found to be dose- and time-dependent. Nidulin also enhanced insulin- and metformin-stimulated glucose uptake. Upregulation of GLUT4 expression and AKT and AMPK phosphorylation were observed with nidulin treatment. Blockage of AKT, but not AMPK, phosphorylation was largely accompanied by diminished glucose uptake. In agreement, nidulin triggered the translocation of GLUT4 to the plasma membrane. Importantly, nidulin elevated glucose uptake associated with increased AKT phosphorylation in insulin-resistant adipocytes. Taken together, nidulin could stimulate glucose uptake mainly through AKT-dependent GLUT4 translocation, serving as a seed compound in drug discovery for type 2 diabetes.
Collapse
Affiliation(s)
- Kanittha Chantarasakha
- National
Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency
(NSTDA), 111 Thailand
Science Park, Phahonyothin Road, Klong Luang, Pathumthani 12120, Thailand
| | - Arunrat Yangchum
- National
Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency
(NSTDA), 111 Thailand
Science Park, Phahonyothin Road, Klong Luang, Pathumthani 12120, Thailand
| | - Masahiko Isaka
- National
Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency
(NSTDA), 111 Thailand
Science Park, Phahonyothin Road, Klong Luang, Pathumthani 12120, Thailand
| | - Surapun Tepaamorndech
- Department
of Microbiology, Faculty of Medicine, Chulalongkorn
University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| |
Collapse
|
3
|
Ramos SV, Distefano G, Lui LY, Cawthon PM, Kramer P, Sipula IJ, Bello FM, Mau T, Jurczak MJ, Molina AJ, Kershaw EE, Marcinek DJ, Shankland E, Toledo FG, Newman AB, Hepple RT, Kritchevsky SB, Goodpaster BH, Cummings SR, Coen PM. Role of Cardiorespiratory Fitness and Mitochondrial Oxidative Capacity in Reduced Walk Speed of Older Adults With Diabetes. Diabetes 2024; 73:1048-1057. [PMID: 38551899 PMCID: PMC11189829 DOI: 10.2337/db23-0827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 03/26/2024] [Indexed: 04/09/2024]
Abstract
Cardiorespiratory fitness and mitochondrial oxidative capacity are associated with reduced walking speed in older adults, but their impact on walking speed in older adults with diabetes has not been clearly defined. We examined differences in cardiorespiratory fitness and skeletal muscle mitochondrial oxidative capacity between older adults with and without diabetes, as well as determined their relative contribution to slower walking speed in older adults with diabetes. Participants with diabetes (n = 159) had lower cardiorespiratory fitness and mitochondrial respiration in permeabilized fiber bundles compared with those without diabetes (n = 717), following adjustments for covariates including BMI, chronic comorbid health conditions, and physical activity. Four-meter and 400-m walking speeds were slower in those with diabetes. Mitochondrial oxidative capacity alone or combined with cardiorespiratory fitness mediated ∼20-70% of the difference in walking speed between older adults with and without diabetes. Additional adjustments for BMI and comorbidities further explained the group differences in walking speed. Cardiorespiratory fitness and skeletal muscle mitochondrial oxidative capacity contribute to slower walking speeds in older adults with diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
| | | | - Li-Yung Lui
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Peggy M. Cawthon
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Philip Kramer
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Ian J. Sipula
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Fiona M. Bello
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Michael J. Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Anthony J. Molina
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Erin E. Kershaw
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - David J. Marcinek
- Department of Radiology, University of Washington School of Medicine, Seattle, WA
| | - Eric Shankland
- Department of Radiology, University of Washington School of Medicine, Seattle, WA
| | - Frederico G.S. Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Anne B. Newman
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Russell T. Hepple
- Department of Physical Therapy, University of Florida, Gainesville, FL
| | - Stephen B. Kritchevsky
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Steven R. Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA
| | - Paul M. Coen
- Translational Research Institute, AdventHealth, Orlando, FL
| |
Collapse
|
4
|
Bubak MP, Davidyan A, O'Reilly CL, Mondal SA, Keast J, Doidge SM, Borowik AK, Taylor ME, Volovičeva E, Kinter MT, Britton SL, Koch LG, Stout MB, Lewis TL, Miller BF. Metformin treatment results in distinctive skeletal muscle mitochondrial remodeling in rats with different intrinsic aerobic capacities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582957. [PMID: 38496648 PMCID: PMC10942369 DOI: 10.1101/2024.03.01.582957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The rationale for the use of metformin as a treatment to slow aging was largely based on data collected from metabolically unhealthy individuals. For healthspan extension metformin will also be used in periods of good health. To understand potential context specificity of metformin treatment on skeletal muscle, we used a rat model (HCR/LCR) with a divide in intrinsic aerobic capacity. Outcomes of metformin treatment differed based on baseline intrinsic mitochondrial function, oxidative capacity of the muscle (gastroc vs soleus), and the mitochondrial population (IMF vs SS). Metformin caused lower ADP-stimulated respiration in LCRs, with less of a change in HCRs. However, a washout of metformin resulted in an unexpected doubling of respiratory capacity in HCRs. These improvements in respiratory capacity were accompanied by mitochondrial remodeling that included increases in protein synthesis and changes in morphology. Our findings raise questions about whether the positive findings of metformin treatment are broadly applicable.
Collapse
|
5
|
Zunica ERM, Heintz EC, Dantas WS, Hebert RC, Tanksley M, Beyl RA, Mader EC, Kirwan JP, Axelrod CL, Singh P. Effects of metformin on glucose metabolism and mitochondrial function in patients with obstructive sleep apnea: A pilot randomized trial. Physiol Rep 2024; 12:e15948. [PMID: 38346816 PMCID: PMC10861357 DOI: 10.14814/phy2.15948] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
Obstructive sleep apnea (OSA) is associated with increased risk for diabetes, and standard treatment with positive airway pressure (PAP) device shows inconsistent effects on glucose metabolism. Metformin is known to treat and prevent diabetes, but its effects on skeletal muscle mitochondrial function are not completely understood. Here, we evaluate the effects of metformin on glucose metabolism and skeletal muscle mitochondrial function in patients with OSA. Sixteen adults with obesity (50.9 ± 6.7 years, BMI: 36.5 ± 2.9 kg/m2 ) and moderate-to-severe OSA were provided with PAP treatment and randomized to 3 months of placebo (n = 8) or metformin (n = 8) treatment in a double-blind parallel-group design. Whole body glucose metabolism was determined by oral glucose tolerance test. A skeletal muscle biopsy was obtained to evaluate mitochondrial respiratory capacity and expression of proteins related to mitochondrial dynamics and energy metabolism. Whole body insulin-sensitivity (Matsuda index) did not change in metformin or placebo treated groups. However, metformin treatment prevented increases in insulin release relative to placebo during follow-up. Insulin area under the curve (AUC) and insulin to glucose AUC ratio increased in placebo but remained unchanged with metformin. Furthermore, metformin treatment improved skeletal muscle mitochondrial respiratory capacity and dynamics relative to placebo. Metformin treatment prevented the decline in whole body glucose homeostasis and skeletal muscle mitochondrial function in patients with moderate to severe OSA. Patients with OSA may benefit from the addition of metformin to prevent diabetes.
Collapse
Affiliation(s)
- Elizabeth R. M. Zunica
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Elizabeth C. Heintz
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Wagner S. Dantas
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - R. Caitlin Hebert
- Translational Physiology LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - MaKayla Tanksley
- Sleep and Cardiometabolic Health LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Robbie A. Beyl
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Edward C. Mader
- Louisiana State University Health Science CenterNew OrleansLouisianaUSA
| | - John P. Kirwan
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Christopher L. Axelrod
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Prachi Singh
- Sleep and Cardiometabolic Health LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| |
Collapse
|
6
|
Tyagi S, Mani S. Combined Administration of Metformin and Vitamin D: A Futuristic Approach for Management of Hyperglycemia. Cardiovasc Hematol Agents Med Chem 2024; 22:258-275. [PMID: 37929731 DOI: 10.2174/0118715257261643231018102928] [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: 06/09/2023] [Revised: 08/28/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
Diabetes is a series of metabolic disorders that can be categorized into three types depending on different aspects associated with age at onset, intensity of insulin resistance, and beta- cell dysfunction: Type 1 and 2 Diabetes, and Gestational Diabetes Mellitus. Type 2 Diabetes Mellitus (T2DM) has recently been found to account for more than 85% of diabetic cases. The current review intends to raise awareness among clinicians/researchers that combining vitamin D3 with metformin may pave the way for better T2DM treatment and management. An extensive literature survey was performed to analyze vitamin D's role in regulating insulin secretion, their action on the target cells and thus maintaining the normal glucose level. On the other side, the anti-hyperglycemic effect of metformin as well as its detailed mechanism of action was also studied. Interestingly both compounds are known to exhibit the antioxidant effect too. Literature supporting the correlation between diabetic phenotypes and deficiency of vitamin D was also explored further. To thoroughly understand the common/overlapping pathways responsible for the antidiabetic as well as antioxidant nature of metformin and vitamin D3, we compared their antihyperglycemic and antioxidant activities. With this background, we are proposing the hypothesis that it would be of great interest if these two compounds could work in synergy to better manage the condition of T2DM and associated disorders.
Collapse
Affiliation(s)
- Sakshi Tyagi
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Shalini Mani
- Centre for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| |
Collapse
|
7
|
Ramos SV, Distefano G, Lui LY, Cawthon PM, Kramer P, Sipula IJ, Bello FM, Mau T, Jurczak MJ, Molina AJ, Kershaw EE, Marcinek DJ, Toledo FGS, Newman AB, Hepple RT, Kritchevsky SB, Goodpaster BH, Cummings SR, Coen PM. Role of Cardiorespiratory Fitness and Mitochondrial Energetics in Reduced Walk Speed of Older Adults with Diabetes in the Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.03.23297992. [PMID: 37986814 PMCID: PMC10659460 DOI: 10.1101/2023.11.03.23297992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Rationale Cardiorespiratory fitness and mitochondrial energetics are associated with reduced walking speed in older adults. The impact of cardiorespiratory fitness and mitochondrial energetics on walking speed in older adults with diabetes has not been clearly defined. Objective To examine differences in cardiorespiratory fitness and skeletal muscle mitochondrial energetics between older adults with and without diabetes. We also assessed the contribution of cardiorespiratory fitness and skeletal muscle mitochondrial energetics to slower walking speed in older adults with diabetes. Findings Participants with diabetes had lower cardiorespiratory fitness and mitochondrial energetics when compared to those without diabetes, following adjustments for covariates including BMI, chronic comorbid health conditions, and physical activity. 4-m and 400-m walking speeds were slower in those with diabetes. Mitochondrial oxidative capacity alone or combined with cardiorespiratory fitness mediated ∼20-70% of the difference in walk speed between older adults with and without diabetes. Further adjustments of BMI and co-morbidities further explained the group differences in walk speed. Conclusions Skeletal muscle mitochondrial energetics and cardiorespiratory fitness contribute to slower walking speeds in older adults with diabetes. Cardiorespiratory fitness and mitochondrial energetics may be therapeutic targets to maintain or improve mobility in older adults with diabetes. ARTICLE HIGHLIGHTS Why did we undertake this study? To determine if mitochondrial energetics and cardiorespiratory fitness contribute to slower walking speed in older adults with diabetes. What is the specific question(s) we wanted to answer? Are mitochondrial energetics and cardiorespiratory fitness in older adults with diabetes lower than those without diabetes? How does mitochondrial energetics and cardiorespiratory fitness impact walking speed in older adults with diabetes? What did we find? Mitochondrial energetics and cardiorespiratory fitness were lower in older adults with diabetes compared to those without diabetes, and energetics, and cardiorespiratory fitness, contributed to slower walking speed in those with diabetes. What are the implications of our findings? Cardiorespiratory fitness and mitochondrial energetics may be key therapeutic targets to maintain or improve mobility in older adults with diabetes.
Collapse
|
8
|
Maurer J, Zhao X, Irmler M, Gudiksen A, Pilmark NS, Li Q, Goj T, Beckers J, Hrabě de Angelis M, Birkenfeld AL, Peter A, Lehmann R, Pilegaard H, Karstoft K, Xu G, Weigert C. Redox state and altered pyruvate metabolism contribute to a dose-dependent metformin-induced lactate production of human myotubes. Am J Physiol Cell Physiol 2023; 325:C1131-C1143. [PMID: 37694284 PMCID: PMC10635655 DOI: 10.1152/ajpcell.00186.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/01/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
Metformin-induced glycolysis and lactate production can lead to acidosis as a life-threatening side effect, but slight increases in blood lactate levels in a physiological range were also reported in metformin-treated patients. However, how metformin increases systemic lactate concentrations is only partly understood. Because human skeletal muscle has a high capacity to produce lactate, the aim was to elucidate the dose-dependent regulation of metformin-induced lactate production and the potential contribution of skeletal muscle to blood lactate levels under metformin treatment. This was examined by using metformin treatment (16-776 μM) of primary human myotubes and by 17 days of metformin treatment in humans. As from 78 µM, metformin induced lactate production and secretion and glucose consumption. Investigating the cellular redox state by mitochondrial respirometry, we found metformin to inhibit the respiratory chain complex I (776 µM, P < 0.01) along with decreasing the [NAD+]:[NADH] ratio (776 µM, P < 0.001). RNA sequencing and phospho-immunoblot data indicate inhibition of pyruvate oxidation mediated through phosphorylation of the pyruvate dehydrogenase (PDH) complex (39 µM, P < 0.01). On the other hand, in human skeletal muscle, phosphorylation of PDH was not altered by metformin. Nonetheless, blood lactate levels were increased under metformin treatment (P < 0.05). In conclusion, the findings suggest that metformin-induced inhibition of pyruvate oxidation combined with altered cellular redox state shifts the equilibrium of the lactate dehydrogenase (LDH) reaction leading to a dose-dependent lactate production in primary human myotubes.NEW & NOTEWORTHY Metformin shifts the equilibrium of lactate dehydrogenase (LDH) reaction by low dose-induced phosphorylation of pyruvate dehydrogenase (PDH) resulting in inhibition of pyruvate oxidation and high dose-induced increase in NADH, which explains the dose-dependent lactate production of differentiated human skeletal muscle cells.
Collapse
Affiliation(s)
- Jennifer Maurer
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Xinjie Zhao
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, The Chinese Academy of Sciences, Dalian, China
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Munich, Neuherberg, Germany
| | - Anders Gudiksen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Nanna S Pilmark
- Centre for Physical Activity Research (CFAS), Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Qi Li
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, The Chinese Academy of Sciences, Dalian, China
| | - Thomas Goj
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, Technical University of Munich, Freising, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, Technical University of Munich, Freising, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Munich, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine IV, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Peter
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Munich, University of Tübingen, Tübingen, Germany
| | - Rainer Lehmann
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Munich, University of Tübingen, Tübingen, Germany
| | - Henriette Pilegaard
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Karstoft
- Centre for Physical Activity Research (CFAS), Rigshospitalet, University Hospital of Copenhagen, Copenhagen, Denmark
- Department of Clinical Pharmacology, Bispebjerg and Fredriksberg Hospital, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Guowang Xu
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, The Chinese Academy of Sciences, Dalian, China
| | - Cora Weigert
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases, Helmholtz Munich, University of Tübingen, Tübingen, Germany
| |
Collapse
|
9
|
Abbadessa G, Maniscalco E, Grasso L, Popara J, Di Scipio F, Franco F, Mancardi D, Pigozzi F, Borrione P, Berta GN, Racca S. Metformin Protects Rat Skeletal Muscle from Physical Exercise-Induced Injury. Biomedicines 2023; 11:2334. [PMID: 37760776 PMCID: PMC10525561 DOI: 10.3390/biomedicines11092334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 07/26/2023] [Accepted: 08/16/2023] [Indexed: 09/29/2023] Open
Abstract
Metformin (Met) is a drug commonly prescribed in type 2 diabetes mellitus. Its efficacy is due to the suppression of hepatic gluconeogenesis, enhancement of peripheral glucose uptake and lower glucose absorption by the intestine. Recent studies have reported Met efficacy in other clinical applications, such as age-related diseases. Despite the wide clinical use of Met, its mechanism of action on muscle and its effect on muscle performance are unclear. We investigated the effects of Met combined with training on physical performance (PP) in healthy rats receiving Met for 8 weeks while undergoing daily moderate exercise. We evaluated the following: PP through graded endurance exercise test performed before the beginning of the training protocol and 48 h before the end of the training period; blood ALT, AST, LDH and CK-MB levels in order to address muscle damage; and several blood and muscle myokines and the expression of factors believed to be involved in muscle adaptation to exercise. Our data demonstrate that Met does not improve the positive effects of exercise on performance, although it protects myocytes from exercise-induced damage. Moreover, given that Met positively affects exercise-induced muscle adaptation, our data support the idea of the therapeutic application of Met when muscle function and structure are compromised.
Collapse
Affiliation(s)
- Giuliana Abbadessa
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Eleonora Maniscalco
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Loredana Grasso
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Jasmin Popara
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Federica Di Scipio
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Francesco Franco
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Daniele Mancardi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.P.); (P.B.)
| | - Paolo Borrione
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy; (F.P.); (P.B.)
| | - Giovanni Nicolao Berta
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| | - Silvia Racca
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (E.M.); (L.G.); (J.P.); (F.D.S.); (F.F.); (D.M.); (S.R.)
| |
Collapse
|
10
|
Zhou J, Shi Y, Yang C, Lu S, Zhao L, Liu X, Zhou D, Luo L, Yin Z. γ-glutamylcysteine alleviates insulin resistance and hepatic steatosis by regulating adenylate cyclase and IGF-1R/IRS1/PI3K/Akt signaling pathways. J Nutr Biochem 2023:109404. [PMID: 37311491 DOI: 10.1016/j.jnutbio.2023.109404] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/15/2023]
Abstract
Type 2 diabetes mellitus (T2DM), a complex metabolism disease, which was characterized by metabolic disorders including hyperglycemia, has become a major health problem due to the increasing prevalence worldwide. γ-glutamylcysteine (γ-GC) as an immediate precursor of glutathione (GSH) was originally used for the treatment of sepsis, inflammation bowel disease, and senescence. Here, we evaluated the capacity of γ-GC on diabetes-related metabolic parameters in db/db mice and insulin resistance (IR) amelioration in cells induced by palmitic acid (PA). Our data suggested that γ-GC treatment decreased body weight, reduced adipose tissue size, ameliorated ectopic fat deposition in liver, increased the GSH content in liver, improved glucose control and other diabetes-related metabolic parameters in vivo. Moreover, in vitro experiments showed that γ-GC could maintain the balance of free fatty acids (FFAs) and glucose uptake through regulating the translocation of CD36 and GLUT4 from cytoplasm to plasma membrane. Furthermore, our finding also provided evidence that γ-GC could activate Akt not only via adenylate cyclase (AC)/cAMP/PI3K signaling pathway, but also via IGF-1R/IRS1/PI3K signaling pathway to improve IR and hepatic steatosis. Blocking either of two signaling pathways could not activate Akt activation induced by γ-GC. This unique characteristic ensures the important role of γ-GC in glucose metabolism. Collectively, these results suggested that γ-GC could serve as a candidate dipeptide for the treatment of T2DM and related chronic diabetic complications via activating AC and IGF-1R/IRS1/PI3K/Akt signaling pathways to regulate CD36 and GLUT4 trafficking.
Collapse
Affiliation(s)
- Jinyi Zhou
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Yingying Shi
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Chen Yang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Shuai Lu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lishuang Zhao
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Xianli Liu
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Da Zhou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China.
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
11
|
Metformin Attenuates Slow-to-Fast Fiber Shift and Proteolysis Markers Increase in Rat Soleus after 7 Days of Rat Hindlimb Unloading. Int J Mol Sci 2022; 24:ijms24010503. [PMID: 36613942 PMCID: PMC9820761 DOI: 10.3390/ijms24010503] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Muscle unloading leads to signaling alterations that cause muscle atrophy and weakness. The cellular energy sensor AMPK can regulate myofiber-type shift, calcium-dependent signaling and ubiquitin-proteasome system markers. We hypothesized that the prevention of p-AMPK downregulation during the first week of muscle unloading would impede atrophy development and the slow-to-fast shift of soleus muscle fibers, and the aim of the study was to test this hypothesis. Thirty-two male Wistar rats were randomly assigned to four groups: placebo control (C), control rats treated with metformin (C + M), 7 days of hindlimb suspension (HS) + placebo (7HS), and 7 days of HS + metformin administration (7HS + M). In the soleus of the 7HS rats, we detected a slow-to-fast fiber-type shift as well as a significant downregulation of MEF-2D and p300 in the nuclei. In the 7HS group, we also found decreases in p-ACC (AMPK target) protein level and in the expression of E3 ubiquitin ligases and p-CaMK II protein level vs. the C group. The 7-day metformin treatment for soleus muscle unloading (1) prevented slow-to-fast fiber-type shift; (2) counteracted changes in the p-ACC protein level; (3) hindered changes in the nuclear protein level of the slow myosin expression activators MEF-2D and p300, but did not affect NFATc1 signaling; and (4) attenuated the unloading-induced upregulation of MuRF-1, atrogin-1, ubiquitin and myostatin mRNA expression, but did not prevent soleus muscle atrophy. Thus, metformin treatment during muscle disuse could be useful to prevent the decrease in the percentage of slow-type fatigue-resistant muscle fibers.
Collapse
|
12
|
Bottoni P, Gionta G, Scatena R. Remarks on Mitochondrial Myopathies. Int J Mol Sci 2022; 24:ijms24010124. [PMID: 36613565 PMCID: PMC9820309 DOI: 10.3390/ijms24010124] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Mitochondrial myopathies represent a heterogeneous group of diseases caused mainly by genetic mutations to proteins that are related to mitochondrial oxidative metabolism. Meanwhile, a similar etiopathogenetic mechanism (i.e., a deranged oxidative phosphorylation and a dramatic reduction of ATP synthesis) reveals that the evolution of these myopathies show significant differences. However, some physiological and pathophysiological aspects of mitochondria often reveal other potential molecular mechanisms that could have a significant pathogenetic role in the clinical evolution of these disorders, such as: i. a deranged ROS production both in term of signaling and in terms of damaging molecules; ii. the severe modifications of nicotinamide adenine dinucleotide (NAD)+/NADH, pyruvate/lactate, and α-ketoglutarate (α-KG)/2- hydroxyglutarate (2-HG) ratios. A better definition of the molecular mechanisms at the basis of their pathogenesis could improve not only the clinical approach in terms of diagnosis, prognosis, and therapy of these myopathies but also deepen the knowledge of mitochondrial medicine in general.
Collapse
Affiliation(s)
- Patrizia Bottoni
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giulia Gionta
- Dipartimento Scienze Anatomiche Istologiche Medico Legali e dell’Apparato Locomotore—Sezione di Anatomia Umana, Università La Sapienza di Roma, Via Alfonso Borelli 50, 00161 Rome, Italy
| | - Roberto Scatena
- Dipartimento di Medicina di Laboratorio, Madre Giuseppina Vannini Hospital, Via di Acqua Bullicante 4, 00177 Rome, Italy
- Correspondence:
| |
Collapse
|
13
|
Erickson ML, Allen JM, Beavers DP, Collins LM, Davidson KW, Erickson KI, Esser KA, Hesselink MKC, Moreau KL, Laber EB, Peterson CA, Peterson CM, Reusch JE, Thyfault JP, Youngstedt SD, Zierath JR, Goodpaster BH, LeBrasseur NK, Buford TW, Sparks LM. Understanding heterogeneity of responses to, and optimizing clinical efficacy of, exercise training in older adults: NIH NIA Workshop summary. GeroScience 2022; 45:569-589. [PMID: 36242693 PMCID: PMC9886780 DOI: 10.1007/s11357-022-00668-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 02/03/2023] Open
Abstract
Exercise is a cornerstone of preventive medicine and a promising strategy to intervene on the biology of aging. Variation in the response to exercise is a widely accepted concept that dates back to the 1980s with classic genetic studies identifying sequence variations as modifiers of the VO2max response to training. Since that time, the literature of exercise response variance has been populated with retrospective analyses of existing datasets that are limited by a lack of statistical power from technical error of the measurements and small sample sizes, as well as diffuse outcomes, very few of which have included older adults. Prospective studies that are appropriately designed to interrogate exercise response variation in key outcomes identified a priori and inclusive of individuals over the age of 70 are long overdue. Understanding the underlying intrinsic (e.g., genetics and epigenetics) and extrinsic (e.g., medication use, diet, chronic disease) factors that determine robust versus poor responses to various exercise factors will be used to improve exercise prescription to target the pillars of aging and optimize the clinical efficacy of exercise training in older adults. This review summarizes the proceedings of the NIA-sponsored workshop entitled, "Understanding Heterogeneity of Responses to, and Optimizing Clinical Efficacy of, Exercise Training in Older Adults" and highlights the importance and current state of exercise response variation research, particularly in older adults, prevailing challenges, and future directions.
Collapse
Affiliation(s)
- Melissa L Erickson
- Translational Research Institute, AdventHealth, 301 E Princeton St, Orlando, FL, 32804, USA
| | - Jacob M Allen
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Daniel P Beavers
- Department of Statistical Sciences, Wake Forest University, Winston-Salem, NC, USA
| | - Linda M Collins
- Department of Social and Behavioral Sciences, New York University, New York, NY, USA
| | - Karina W Davidson
- Institute of Health System Science, Feinstein Institutes for Medical Research, Northwell Health, New York, NY, USA
| | - Kirk I Erickson
- Translational Research Institute, AdventHealth, 301 E Princeton St, Orlando, FL, 32804, USA
| | - Karyn A Esser
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Kerrie L Moreau
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eric B Laber
- Department of Statistical Sciences, Duke University, Durham, NC, USA
| | - Charlotte A Peterson
- Center for Muscle Biology, College of Health Sciences, University of Kentucky, Lexington, KY, USA
| | - Courtney M Peterson
- Department of Nutritional Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jane E Reusch
- Department of Medicine, Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KN, USA
| | - Shawn D Youngstedt
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Juleen R Zierath
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Bret H Goodpaster
- Translational Research Institute, AdventHealth, 301 E Princeton St, Orlando, FL, 32804, USA
| | - Nathan K LeBrasseur
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Thomas W Buford
- Department of Medicine, University of Alabama at Birmingham, 1313 13th St. S., Birmingham, AL, 35244, USA.
- Birmingham/Atlanta VA GRECC, Birmingham VA Medical Center, Birmingham, AL, USA.
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, 301 E Princeton St, Orlando, FL, 32804, USA.
| |
Collapse
|
14
|
Avram VF, Merce AP, Hâncu IM, Bătrân AD, Kennedy G, Rosca MG, Muntean DM. Impairment of Mitochondrial Respiration in Metabolic Diseases: An Overview. Int J Mol Sci 2022; 23:8852. [PMID: 36012137 PMCID: PMC9408127 DOI: 10.3390/ijms23168852] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial dysfunction has emerged as a central pathomechanism in the setting of obesity and diabetes mellitus, linking these intertwined pathologies that share insulin resistance as a common denominator. High-resolution respirometry (HRR) is a state-of-the-art research method currently used to study mitochondrial respiration and its impairment in health and disease. Tissue samples, cells or isolated mitochondria are exposed to various substrate-uncoupler-inhibitor-titration protocols, which allows the measurement and calculation of several parameters of mitochondrial respiration. In this review, we discuss the alterations of mitochondrial bioenergetics in the main dysfunctional organs that contribute to the development of the obese and diabetic phenotypes in both animal models and human subjects. Herein we review data regarding the impairment of oxidative phosphorylation as integrated mitochondrial function assessed by means of HRR. We acknowledge the critical role of this method in determining the alterations in oxidative phosphorylation occurring in the early stages of metabolic pathologies. We conclude that there is a mutual two-way relationship between mitochondrial dysfunction and insulin insensitivity that characterizes these diseases.
Collapse
Affiliation(s)
- Vlad Florian Avram
- Department VII Internal Medicine—Diabetes, Nutrition and Metabolic Diseases, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Molecular Research in Nephrology and Vascular Disease, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Adrian Petru Merce
- Doctoral School Medicine—Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Iasmina Maria Hâncu
- Doctoral School Medicine—Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Alina Doruța Bătrân
- Doctoral School Medicine—Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| | - Gabrielle Kennedy
- Department of Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI 48858, USA
| | - Mariana Georgeta Rosca
- Department of Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI 48858, USA
| | - Danina Mirela Muntean
- Center for Translational Research and Systems Medicine, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
- Department III Functional Sciences—Pathophysiology, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timișoara, Romania
| |
Collapse
|
15
|
Pavlovic K, Krako Jakovljevic N, Isakovic AM, Ivanovic T, Markovic I, Lalic NM. Therapeutic vs. Suprapharmacological Metformin Concentrations: Different Effects on Energy Metabolism and Mitochondrial Function in Skeletal Muscle Cells in vitro. Front Pharmacol 2022; 13:930308. [PMID: 35873556 PMCID: PMC9299382 DOI: 10.3389/fphar.2022.930308] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022] Open
Abstract
Metformin is an oral antidiabetic agent that has been widely used in clinical practice for over 60 years, and is currently the most prescribed antidiabetic drug worldwide. However, the molecular mechanisms of metformin action in different tissues are still not completely understood. Although metformin-induced inhibition of mitochondrial respiratory chain Complex I and activation of AMP-activated protein kinase have been observed in many studies, published data is inconsistent. Furthermore, metformin concentrations used for in vitro studies and their pharmacological relevance are a common point of debate. The aim of this study was to explore the effects of different metformin concentrations on energy metabolism and activity of relevant signaling pathways in C2C12 muscle cells in vitro. In order to determine if therapeutic metformin concentrations have an effect on skeletal muscle cells, we used micromolar metformin concentrations (50 µM), and compared the effects with those of higher, millimolar concentrations (5 mM), that have already been established to affect mitochondrial function and AMPK activity. We conducted all experiments in conditions of high (25 mM) and low glucose (5.5 mM) concentration, in order to discern the role of glucose availability on metformin action. According to our results, micromolar metformin treatment did not cause Complex I inhibition nor AMPK activation. Also, cells cultured in low glucose medium were more sensitive to Complex I inhibition, mitochondrial membrane depolarization and AMPK activation by millimolar metformin, but cells cultured in high glucose medium were more prone to induction of ROS production. In conclusion, even though suprapharmacological metformin concentrations cause Complex I inhibition and AMPK activation in skeletal muscle cells in vitro, therapeutic concentrations cause no such effect. This raises the question if these mechanisms are relevant for therapeutic effects of metformin in skeletal muscle.
Collapse
Affiliation(s)
- Kasja Pavlovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| | - Nina Krako Jakovljevic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| | - Andjelka M Isakovic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Tijana Ivanovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivanka Markovic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
| | - Nebojsa M Lalic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade, Serbia
| |
Collapse
|
16
|
Moreno‐Cabañas A, Morales‐Palomo F, Alvarez‐Jimenez L, Ortega JF, Mora‐Rodriguez R. Effects of chronic metformin treatment on training adaptations in men and women with hyperglycemia: A prospective study. Obesity (Silver Spring) 2022; 30:1219-1230. [PMID: 35578807 PMCID: PMC9321693 DOI: 10.1002/oby.23410] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
OBJECTIVE This study aimed to determine whether chronic metformin use interferes with the improvements in insulin resistance (IR) and cardiorespiratory fitness with aerobic training in people with hyperglycemia and metabolic syndrome (MetS). METHODS A total of 63 middle-aged (53 [7] years) individuals with MetS and obesity (BMI = 32.8 [4.5] kg/m2 ) completed 16 weeks of supervised high-intensity interval training (3 d/wk, 43 min/session). Participants were either taking metformin (EXER+MET; n = 29) or were free of any pharmacological treatment for their MetS factors (EXER; n = 34). Groups were similar in their initial cardiorespiratory fitness (maximal oxygen uptake [VO2MAX ]), age, percentage of women, BMI, and MetS factors (z score). The effects of exercise training on IR (homeostatic model assessment of insulin resistance [HOMA-IR]), MetS z score, VO2MAX , maximal fat oxidation during exercise, and maximal aerobic power output were measured. RESULTS Fasting insulin and HOMA-IR decreased similarly in both groups with training (EXER+MET: -4.3% and -10.6%; EXER: -5.3% and -14.5%; p value for time = 0.005). However, metformin use reduced VO2MAX improvements by half (i.e., EXER+MET: 12.7%; EXER: 25.3%; p value for time × group = 0.012). Maximal fat oxidation during exercise increased similarly in both groups (EXER+MET: 20.7%; EXER: 25.3%; p value for time = 0.040). VO2MAX gains were not associated with HOMA-IR reductions (EXER+MET: r = -0.098; p = 0.580; EXER: r = -0.255; p = 0.182). CONCLUSIONS Metformin use was associated with attenuated VO2MAX improvements but did not affect fasting IR reductions with aerobic training in individuals with hyperglycemia and high cardiovascular risk (i.e., MetS).
Collapse
|
17
|
Feng L, Gao Q, Hu K, Wu M, Wang Z, Chen F, Mei F, Zhao L, Ma B. Prevalence and Risk Factors of Sarcopenia in Patients With Diabetes: A Meta-analysis. J Clin Endocrinol Metab 2022; 107:1470-1483. [PMID: 34904651 DOI: 10.1210/clinem/dgab884] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT The prevalence of sarcopenia in patients with diabetes is 3 times higher than that in patients without diabetes and is associated with a poor prognosis. OBJECTIVE To investigate the global pooled prevalence and risk factors of sarcopenia in patients with diabetes. DATA SOURCES Relevant studies published until November 30, 2020, were identified from the PubMed, Embase, Web of Science, WanFang, CNKI, VIP, and CBM databases. STUDY SELECTION Participants with age ≥ 18 years with clinically diagnosed diabetes. Sex and diabetes type were not restricted. DATA EXTRACTION The data were extracted by 2 reviewers independently using a standard data collection form. DATA SYNTHESIS The pooled prevalence of sarcopenia in patients with diabetes was 18% (95% CI, 16-20); subgroup analysis showed that sarcopenia was more prevalent in males than in females, as well as being more prevalent in Asia than in South America and Oceania. Age (odds ratio [OR], 1.10), glycated hemoglobin (HbA1c) (OR = 1.16), visceral fat area (VFA) (OR = 1.03), diabetic nephropathy (OR = 2.54), duration of diabetes (OR = 1.06), and high-sensitivity C-reactive protein (hs-CRP) (OR = 1.33) were risk factors for sarcopenia in patients with diabetes. CONCLUSIONS Sarcopenia was more prevalent in patients with diabetes. Age, HbA1c, VFA, diabetic nephropathy, duration of diabetes, and hs-CRP were the probable risk factors. In the future, medical staff should not only pay attention to the early screening of sarcopenia in high-risk groups, but also provide information on its prevention.
Collapse
Affiliation(s)
- Liyuan Feng
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
| | - Qianqian Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
| | - Kaiyan Hu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
- Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, P.R. China
| | - Mei Wu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
| | - Zhe Wang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
| | - Fei Chen
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
| | - Fan Mei
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
| | - Li Zhao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
| | - Bin Ma
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Evidence-Based Nursing Center, School of Nursing, Lanzhou University, Lanzhou 730000, P.R. China
- Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou 730000, P.R. China
| |
Collapse
|
18
|
D-galactose-induced aging in rats – The effect of metformin on bioenergetics of brain, skeletal muscle and liver. Exp Gerontol 2022; 163:111770. [DOI: 10.1016/j.exger.2022.111770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/18/2022]
|
19
|
Huang J, Pham M, Panenka WJ, Honer WG, Barr AM. Chronic Treatment With Psilocybin Decreases Changes in Body Weight in a Rodent Model of Obesity. Front Psychiatry 2022; 13:891512. [PMID: 35664477 PMCID: PMC9157591 DOI: 10.3389/fpsyt.2022.891512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/27/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND There are currently relatively few effective pharmacological treatments for obesity, and existing ones may be associated with limiting side-effects. In the search for novel anti-obesity agents, drugs that modify central serotonergic systems have historically proven to be effective in promoting weight loss. Psilocin, which is rapidly metabolized from psilocybin, is an agonist at multiple serotonin receptors. In the present study we assessed the effects of psilocybin and a positive control (metformin) on changes in body weight in a rat model of obesity. METHODS Five groups of adult male rats were pre-conditioned with a cafeteria diet until obese (>600 g) and then treated with either psilocybin (0.1, 1, or 5 mg/kg, i.p.), metformin (300 mg/kg, p.o.) or vehicle control. Treatments were for 27 consecutive weekdays, and body weights and high calorie food intake were recorded daily. Fasting glucose levels were recorded after 11 days of treatment. At the end of treatment rats completed a glucose tolerance test, and multiple fat pads were dissected out to assess adiposity. RESULTS The medium dose psilocybin group had to be terminated from the study prematurely. Both the low and high dose psilocybin groups caused a significant decrease in changes in body weight compared to controls. The metformin group produced a greater decrease in change in body weight than either psilocybin groups or controls. Both high dose psilocybin and metformin decreased consumption of the high calorie diet, and exhibited decreased central adiposity. CONCLUSION Psilocybin demonstrated modest but significant effects on weight gain. Further study is recommended.
Collapse
Affiliation(s)
- Joyce Huang
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Michelle Pham
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - William J Panenka
- Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,British Columbia Mental Health & Substance Use Services Research Institute, Vancouver, BC, Canada
| | - William G Honer
- Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,British Columbia Mental Health & Substance Use Services Research Institute, Vancouver, BC, Canada
| | - Alasdair M Barr
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,British Columbia Mental Health & Substance Use Services Research Institute, Vancouver, BC, Canada
| |
Collapse
|
20
|
Kumari S, Bubak MT, Schoenberg HM, Davidyan A, Elliehausen CJ, Kuhn KG, VanWagoner TM, Karaman R, Scofield RH, Miller BF, Konopka AR. Antecedent Metabolic Health and Metformin (ANTHEM) Aging Study: Rationale and Study Design for a Randomized Controlled Trial. J Gerontol A Biol Sci Med Sci 2021; 77:2373-2377. [PMID: 34865016 PMCID: PMC9799202 DOI: 10.1093/gerona/glab358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Indexed: 01/21/2023] Open
Abstract
The antidiabetic medication metformin has been proposed to be the first drug tested to target aging and extend healthspan in humans. While there is extensive epidemiological support for the health benefits of metformin in patient populations, it is not clear if these protective effects apply to those free of age-related disease. Our previous data in older adults without diabetes suggest a dichotomous change in insulin sensitivity and skeletal muscle mitochondrial adaptations after metformin treatment when co-prescribed with exercise. Those who entered the study as insulin-sensitive had no change to detrimental effects while those who were insulin-resistant had positive changes. The objective of this clinical trial is to determine if (a) antecedent metabolic health and (b) skeletal muscle mitochondrial remodeling and function mediate the positive or detrimental effects of metformin monotherapy, independent of exercise, on the metabolism and biology of aging. In a randomized, double-blind clinical trial, adults free of chronic disease (n = 148, 40-75 years old) are stratified as either insulin-sensitive or resistant based on homeostatic model assessment of insulin resistance (≤2.2 or ≥2.5) and take 1 500 mg/day of metformin or placebo for 12 weeks. Hyperinsulinemic-euglycemic clamps and skeletal muscle biopsies are performed before and after 12 weeks to assess primary outcomes of peripheral insulin sensitivity and mitochondrial remodeling and function. Findings from this trial will identify clinical characteristics and cellular mechanisms involved in modulating the effectiveness of metformin treatment to target aging that could inform larger Phase 3 clinical trials aimed at testing aging as a treatment indication for metformin. Clinical Trials Registration Number: NCT04264897.
Collapse
Affiliation(s)
| | - Matthew T Bubak
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Hayden M Schoenberg
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA,Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Arik Davidyan
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Christian J Elliehausen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA,Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Katrin G Kuhn
- Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Timothy M VanWagoner
- Oklahoma Shared Clinical and Translational Resources, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Rowan Karaman
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Wisconsin–Madison, Madison, Wisconsin, USA,Division of Endocrinology, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Robert Hal Scofield
- Department of Veterans Affairs Medical Center, Oklahoma City, Oklahoma, USA,Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA,Endocrinology, Diabetes and Metabolism Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - Adam R Konopka
- Address correspondence to: Adam R. Konopka, PhD, Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin–Madison and Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA. E-mail:
| |
Collapse
|
21
|
Ma XY, Chen FQ. Effects of anti-diabetic drugs on sarcopenia: Best treatment options for elderly patients with type 2 diabetes mellitus and sarcopenia. World J Clin Cases 2021; 9:10064-10074. [PMID: 34904076 PMCID: PMC8638038 DOI: 10.12998/wjcc.v9.i33.10064] [Citation(s) in RCA: 6] [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: 07/14/2021] [Revised: 08/22/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Human life expectancy increases as society becomes more developed. This increased life expectancy poses challenges associated with the rapid aging of the population. Sarcopenia, an age-related disease, has become a worldwide health issue. Patients with sarcopenia experience decreases in muscle mass and function, becoming frail and eventually bedridden. Type 2 diabetes mellitus (T2DM) is also a major health issue; the incidence of T2DM increases with aging. T2DM is associated with reduced muscle strength and poor muscle quality and may contribute to acceleration of the aging process, augmenting age-related sarcopenia. Recent studies indicate that elderly patients with diabetes are at an increased risk for sarcopenia. Therefore, these older diabetic patients with sarcopenia need specific anti-diabetic therapies targeting not only glycemic control but also sarcopenia, with the goal of preventing sarcopenia in pre-sarcopenic patients. Presently, various types of hypoglycemic drugs are available, but which hypoglycemic drugs are better suited for geriatric T2DM patients with sarcopenia remains undetermined. In this review, we discuss the association between diabetes and sarcopenia in geriatric patients, and how anti-diabetic drugs may influence sarcopenia outcomes. This review will guide clinical workers in the selection of drugs best suited for this patient population.
Collapse
Affiliation(s)
- Xiao-Yu Ma
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| | - Fen-Qin Chen
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
22
|
Herbst A, Hoang A, Kim C, Aiken JM, McKenzie D, Goldwater DS, Wanagat J. Metformin Treatment in Old Rats and Effects on Mitochondrial Integrity. Rejuvenation Res 2021; 24:434-440. [PMID: 34779265 DOI: 10.1089/rej.2021.0052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Metformin, a commonly used well-tolerated treatment for type 2 diabetes, is being deployed in clinical trials to ameliorate aging in older nondiabetic humans. Concerningly, some experiments in model organisms have suggested that metformin use at old ages shortens life span and is toxic to mitochondria. The demonstrated safety of metformin therapy in humans and the conflicting data from model organisms compelled us to test the hypothesis that metformin treatment would be toxic to older rats. To define an effective dose in 30-month-old hybrid rats, we evaluated two doses of metformin (0.1%, 0.75% of the diet) and treated the rats for 4 months. Body mass decreased at the 0.75% dose. Neither dose affected mortality between 30 and 34 months of age. We assessed mitochondrial integrity by measuring mitochondrial DNA (mtDNA) copy number and deletion mutation frequency, and mitochondrial respiration in skeletal muscle and the heart. In skeletal muscle, we observed no effect of metformin on quadriceps mass, mtDNA copy number, or deletion frequency. In the heart, metformin-treated rats had higher mtDNA copy number, lower cardiac mass, with no change in mtDNA deletion frequency. Metformin treatment resulted in lower mitochondrial complex I-dependent respiration in the heart. We found that, in old rats, metformin did not compromise mtDNA integrity, did not affect mortality, and may have cardiac benefits. These data provide some reassurance that a metformin intervention in aged mammals is not toxic at appropriate doses.
Collapse
Affiliation(s)
- Allen Herbst
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Canada
| | - Austin Hoang
- Division of Geriatrics, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Chiye Kim
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Judd M Aiken
- Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Canada
| | - Debbie McKenzie
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Deena S Goldwater
- Division of Geriatrics, Department of Medicine, UCLA, Los Angeles, California, USA.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California, USA.,Division of Cardiology, Department of Medicine, UCLA, Los Angeles, California, USA
| | - Jonathan Wanagat
- Division of Geriatrics, Department of Medicine, UCLA, Los Angeles, California, USA.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
23
|
Pilmark NS, Oberholzer L, Halling JF, Kristensen JM, Bønding CP, Elkjær I, Lyngbæk M, Elster G, Siebenmann C, Holm NF, Birk JBB, Larsen EL, Meinild-Lundby AK, Wojtaszewski JF, Pilegaard H, Poulsen H, Pedersen BK, Hansen KB, Karstoft K. Skeletal muscle adaptations to exercise are not influenced by metformin treatment in humans: secondary analyses of two randomised, clinical trials. Appl Physiol Nutr Metab 2021; 47:309-320. [PMID: 34784247 DOI: 10.1139/apnm-2021-0194] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Metformin and exercise both improve glycemic control, but in vitro studies have indicated that an interaction between metformin and exercise occurs in skeletal muscle, suggesting a blunting effect of metformin on exercise training adaptations. Two studies (a double-blind, parallel-group, randomized clinical trial conducted in 29 glucose-intolerant individuals and a double-blind, cross-over trial conducted in 15 healthy lean males) were included in this paper. In both studies, the effect of acute exercise +/- metformin treatment on different skeletal muscle variables, previously suggested to be involved in a pharmaco-physiological interaction between metformin and exercise, was assessed. Furthermore, in the parallel-group trial, the effect of 12 weeks of exercise training was assessed. Skeletal muscle biopsies were obtained before and after acute exercise and 12 weeks of exercise training, and mitochondrial respiration, oxidative stress and AMPK activation was determined. Metformin did not significantly affect the effects of acute exercise or exercise training on mitochondrial respiration, oxidative stress or AMPK activation, indicating that the response to acute exercise and exercise training adaptations in skeletal muscle is not affected by metformin treatment. Further studies are needed to investigate whether an interaction between metformin and exercise is present in other tissues, e.g. the gut. Trial registration: ClinicalTrials.gov (NCT03316690 and NCT02951260). Novelty bullets • Metformin does not affect exercise-induced alterations in mitochondrial respiratory capacity in human skeletal muscle • Metformin does not affect exercise-induced alterations in systemic levels of oxidative stress nor emission of reactive oxygen species from human skeletal muscle • Metformin does not affect exercise-induced AMPK activation in human skeletal muscle.
Collapse
Affiliation(s)
- Nanna Skytt Pilmark
- Rigshospitalet, 53146, Centre for Physical Activity Research (CFAS), Copenhagen, Denmark;
| | - Laura Oberholzer
- Center for Physical Activity Research, University Hospital of Copenhagen, Blegdamsvej 9, Copenhagen, Denmark, 2100;
| | - Jens Frey Halling
- university of copenhagen, department of biology, , copenhagen, Denmark;
| | - Jonas M Kristensen
- University of Copenhagen, Denmark, Department of Nutrition, Exercise and Sports,, copenhagen, Denmark;
| | | | - Ida Elkjær
- Center for Physical Activity Research, University Hospital of Copenhagen, Copenhagen, Denmark;
| | - Mark Lyngbæk
- Center for Physical Activity Research, University Hospital of Copenhagen, Copenhagen, Denmark;
| | - Grit Elster
- Center for Physical Activity Research, University Hospital of Copenhagen, Copenhagen, Denmark;
| | - Christoph Siebenmann
- Institute of Mountain Emergency Medicine,, EURAC Research, Bolzano, Italy, bolzano, Italy;
| | - Niels Frederich Holm
- Center for Physical Activity Research, University Hospital of Copenhagen, Copenhagen, Denmark;
| | - Jesper Bratz Bratz Birk
- University of Copenhagen, Denmark, Department of Nutrition, Exercise and Sports,, copenhagen, Denmark;
| | - Emil List Larsen
- Copenhagen University Hospital, 53146, Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, Kobenhavn, Denmark;
| | | | - J F Wojtaszewski
- University of Copenhagen, Denmark, Department of Nutrition, Exercise and Sports,, copenhagen, Denmark;
| | | | - Henrik Poulsen
- Copenhagen University Hospital, 53146, Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, Kobenhavn, Denmark;
| | - Bente Klarlund Pedersen
- Rigshospitalet, 53146, Centre of Inflammation and Metabolism / Centre for Physical Activity Research (CIM/CFAS), København, Denmark;
| | - Katrine Bagge Hansen
- Steno Diabetes Center Copenhagen, 53138, Steno Diabetes Center Copenhagen, Gentofte, Denmark, Gentofte, Denmark;
| | - Kristian Karstoft
- Rigshospitalet, 53146, Centre for Physical Activity Research (CFAS), Blegdamsvej 9, Copenhagen, Denmark, 2100.,Bispebjerg Hospital, 53166, Department of Clinical Pharmacology, Copenhagen, Denmark, 2400;
| |
Collapse
|
24
|
Mora FAA, Musheshe N, Arroyave Ospina JC, Geng Y, Soto JM, Rodrigo JA, Alieva T, Buist-Homan M, Lezoualc'h F, Cheng X, Schmidt M, Moshage H. Metformin protects against diclofenac-induced toxicity in primary rat hepatocytes by preserving mitochondrial integrity via a pathway involving EPAC. Biomed Pharmacother 2021; 143:112072. [PMID: 34464747 DOI: 10.1016/j.biopha.2021.112072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/23/2021] [Accepted: 08/17/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE It has been shown that the antidiabetic drug metformin protects hepatocytes against toxicity by various stressors. Chronic or excessive consumption of diclofenac (DF) - a pain-relieving drug, leads to drug-induced liver injury via a mechanism involving mitochondrial damage and ultimately apoptotic death of hepatocytes. However, whether metformin protects against DF-induced toxicity is unknown. Recently, it was also shown that cAMP elevation is protective against DF-induced apoptotic death in hepatocytes, a protective effect primarily involving the downstream cAMP effector EPAC and preservation of mitochondrial function. This study therefore aimed at investigating whether metformin protects against DF-induced toxicity via cAMP-EPACs. EXPERIMENTAL APPROACH Primary rat hepatocytes were exposed to 400 µmol/L DF. CE3F4 or ESI-O5 were used as EPAC-1 or 2 inhibitors respectively. Apoptosis was measured by caspase-3 activity and necrosis by Sytox green staining. Seahorse X96 assay was used to determine mitochondrial function. Mitochondrial reactive oxygen species (ROS) production was measured using MitoSox, mitochondrial MnSOD expression was determined by immunostaining and mitochondrial morphology (fusion and fission ratio) by 3D refractive index imaging. KEY RESULTS Metformin (1 mmol/L) was protective against DF-induced apoptosis in hepatocytes. This protective effect was EPAC-dependent (mainly EPAC-2). Metformin restored mitochondrial morphology in an EPAC-independent manner. DF-induced mitochondrial dysfunction which was demonstrated by decreased oxygen consumption rate, an increased ROS production and a reduced MnSOD level, were all reversed by metformin in an EPAC-dependent manner. CONCLUSION AND IMPLICATIONS Metformin protects hepatocytes against DF-induced toxicity via cAMP-dependent EPAC-2.
Collapse
Affiliation(s)
- Fabio Alejandro Aguilar Mora
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Nshunge Musheshe
- Deptartment Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen University of Groningen, Groningen, The Netherlands.
| | - Johanna C Arroyave Ospina
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Yana Geng
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Juan M Soto
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - José A Rodrigo
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - Tatiana Alieva
- Department of Optics and Faculty of Physical Sciences, Complutense University of Madrid, Spain.
| | - Manon Buist-Homan
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Frank Lezoualc'h
- Inserm UMR-1048, Institut des Maladies Metaboliques et Cardiovasculaires, Univ Toulouse Paul Sabatier, Toulouse, France.
| | - Xiaodong Cheng
- Department of Integrative Biology & Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Martina Schmidt
- Deptartment Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen University of Groningen, Groningen, The Netherlands.
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
25
|
Skeletal muscle mitochondrial respiration in a model of age-related osteoarthritis is impaired after dietary rapamycin. Exp Gerontol 2021; 155:111579. [PMID: 34601078 DOI: 10.1016/j.exger.2021.111579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 12/24/2022]
Abstract
A decline in skeletal muscle mitochondrial function is associated with the loss of skeletal muscle size and function during knee osteoarthritis (OA). We have recently reported that 12-weeks of dietary rapamycin (Rap, 14 ppm), with or without metformin (Met, 1000 ppm), increased plasma glucose and OA severity in male Dunkin Hartley (DH) guinea pigs, a model of naturally occurring, age-related OA. The purpose of the current study was to determine if increased OA severity after dietary Rap and Rap+Met was accompanied by impaired skeletal muscle mitochondrial function. Mitochondrial respiration and hydrogen peroxide (H2O2) emissions were evaluated in permeabilized muscle fibers via high-resolution respirometry and fluorometry using either a saturating bolus or titration of ADP. Rap and Rap+Met decreased complex I (CI)-linked respiration and tended to increase ADP sensitivity, consistent with previous findings in patients with end-stage OA. The decrease in CI-linked respiration was accompanied with lower CI protein abundance. Rap and Rap+Met did not change mitochondrial H2O2 emissions. There were no differences between mitochondrial function in Rap versus Rap+Met suggesting that Rap was likely driving the change in mitochondrial function. This is the first inquiry into how lifespan extending treatments Rap and Rap+Met can influence skeletal muscle mitochondria in a model of age-related OA. Collectively, our data suggest that Rap with or without Met inhibits CI-linked capacity and increases ADP sensitivity in DH guinea pigs that have greater OA severity.
Collapse
|
26
|
Shoshan-Barmatz V, Anand U, Nahon-Crystal E, Di Carlo M, Shteinfer-Kuzmine A. Adverse Effects of Metformin From Diabetes to COVID-19, Cancer, Neurodegenerative Diseases, and Aging: Is VDAC1 a Common Target? Front Physiol 2021; 12:730048. [PMID: 34671273 PMCID: PMC8521008 DOI: 10.3389/fphys.2021.730048] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin has been used for treating diabetes mellitus since the late 1950s. In addition to its antihyperglycemic activity, it was shown to be a potential drug candidate for treating a range of other diseases that include various cancers, cardiovascular diseases, diabetic kidney disease, neurodegenerative diseases, renal diseases, obesity, inflammation, COVID-19 in diabetic patients, and aging. In this review, we focus on the important aspects of mitochondrial dysfunction in energy metabolism and cell death with their gatekeeper VDAC1 (voltage-dependent anion channel 1) as a possible metformin target, and summarize metformin's effects in several diseases and gut microbiota. We question how the same drug can act on diseases with opposite characteristics, such as increasing apoptotic cell death in cancer, while inhibiting it in neurodegenerative diseases. Interestingly, metformin's adverse effects in many diseases all show VDAC1 involvement, suggesting that it is a common factor in metformin-affecting diseases. The findings that metformin has an opposite effect on various diseases are consistent with the fact that VDAC1 controls cell life and death, supporting the idea that it is a target for metformin.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | | | - Marta Di Carlo
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Anna Shteinfer-Kuzmine
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
27
|
Sartori R, Hagg A, Zampieri S, Armani A, Winbanks CE, Viana LR, Haidar M, Watt KI, Qian H, Pezzini C, Zanganeh P, Turner BJ, Larsson A, Zanchettin G, Pierobon ES, Moletta L, Valmasoni M, Ponzoni A, Attar S, Da Dalt G, Sperti C, Kustermann M, Thomson RE, Larsson L, Loveland KL, Costelli P, Megighian A, Merigliano S, Penna F, Gregorevic P, Sandri M. Perturbed BMP signaling and denervation promote muscle wasting in cancer cachexia. Sci Transl Med 2021; 13:eaay9592. [PMID: 34349036 DOI: 10.1126/scitranslmed.aay9592] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 03/18/2021] [Indexed: 02/05/2023]
Abstract
Most patients with advanced solid cancers exhibit features of cachexia, a debilitating syndrome characterized by progressive loss of skeletal muscle mass and strength. Because the underlying mechanisms of this multifactorial syndrome are incompletely defined, effective therapeutics have yet to be developed. Here, we show that diminished bone morphogenetic protein (BMP) signaling is observed early in the onset of skeletal muscle wasting associated with cancer cachexia in mouse models and in patients with cancer. Cancer-mediated factors including Activin A and IL-6 trigger the expression of the BMP inhibitor Noggin in muscle, which blocks the actions of BMPs on muscle fibers and motor nerves, subsequently causing disruption of the neuromuscular junction (NMJ), denervation, and muscle wasting. Increasing BMP signaling in the muscles of tumor-bearing mice by gene delivery or pharmacological means can prevent muscle wasting and preserve measures of NMJ function. The data identify perturbed BMP signaling and denervation of muscle fibers as important pathogenic mechanisms of muscle wasting associated with tumor growth. Collectively, these findings present interventions that promote BMP-mediated signaling as an attractive strategy to counteract the loss of functional musculature in patients with cancer.
Collapse
Affiliation(s)
- Roberta Sartori
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Adam Hagg
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
- Biomedicine Discovery Institute, Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
| | - Sandra Zampieri
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
- Myology Center, University of Padova, 35122 Padua, Italy
| | - Andrea Armani
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | | | - Laís R Viana
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Structural and Functional Biology, Biology Institute, University of Campinas, Campinas, São Paulo 13083-97, Brazil
| | - Mouna Haidar
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Kevin I Watt
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Hongwei Qian
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Camilla Pezzini
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Pardis Zanganeh
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Anna Larsson
- Theme Cancer, Karolinska University Hospital, Solna 171 76, Sweden
| | - Gianpietro Zanchettin
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Elisa S Pierobon
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Lucia Moletta
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Michele Valmasoni
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Alberto Ponzoni
- Department of Radiology, Padova General Hospital, 35121 Padova, Italy
| | - Shady Attar
- Department of Medicine, University Hospital of Padova, 35121 Padova, Italy
| | - Gianfranco Da Dalt
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Cosimo Sperti
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Monika Kustermann
- Center for Anatomy and Cell Biology, Medical University of Vienna, 1090 Vienna, Austria
| | - Rachel E Thomson
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kate L Loveland
- Centre for Reproductive Health. Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
- Department of Molecular and Translational Sciences, and Anatomy and Developmental Biology, Monash University, VIC 3800, Australia
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Stefano Merigliano
- Department of Surgery, Oncology and Gastroenterology, 3rd Surgical Clinic, University of Padova, 35128 Padua, Italy
| | - Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, 10125 Turin, Italy
| | - Paul Gregorevic
- Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia.
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Biochemistry and Molecular Biology, Monash University, VIC 3800, Australia
- Department of Neurology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Marco Sandri
- Veneto Institute of Molecular Medicine, 35129 Padova, Italy.
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Myology Center, University of Padova, 35122 Padua, Italy
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
28
|
Jin Z, Sun Y, Yang T, Tan L, Lv P, Xu Q, Tao G, Qin S, Lu X, He Q. Nanocapsule-mediated sustained H 2 release in the gut ameliorates metabolic dysfunction-associated fatty liver disease. Biomaterials 2021; 276:121030. [PMID: 34298442 DOI: 10.1016/j.biomaterials.2021.121030] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/14/2021] [Accepted: 07/14/2021] [Indexed: 12/20/2022]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is estimated to affect a quarter of all population and represents a major health threat to all societies. Yet, currently no approved pharmacological treatment is available for MAFLD. H2-rich water has recently been reported to reduce hepatic lipid accumulation in MAFLD patients but its efficacy is limited due to low H2 dosage. Increasing H2 dose may enhance its therapeutic effects but remains technically challenging. In this study, we designed and synthesized a hydrogen nanocapsule by encapsulating ammonia borane into hollow mesoporous silica nanoparticles to achieve ultrahigh and sustained H2 release in the gut. We then investigated its efficacy in treating early-stage MAFLD and other metabolic dysfunctions such as obesity and diabetes. The hydrogen nanocapsule attenuated both diet-induced and genetic mutation induced early-stage MAFLD, obesity, and diabetes in mice, without any tissue toxicity. Mechanistically, we discovered that sustained and ultrahigh H2 supply by hydrogen nanocapsule increased, among other species, the abundance of Akkermansia muciniphila, highlighting reshaped gut microbiota as a potential mechanism of H2 in treating metabolic dysfunctions. Moreover, hepatic transcriptome showed a reprogramed liver metabolism profile with reduced lipid synthesis and increased fatty acid metabolism.
Collapse
Affiliation(s)
- Zhaokui Jin
- Guangdong Provincial Key Laboratory of Immunity and Diseases, Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China; School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Yuan Sun
- Department of Physiology, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China; Department of Pharmacology, College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Tian Yang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Lunbo Tan
- School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China; Department of Physiology, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Peixun Lv
- School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Qingqing Xu
- School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China
| | - Geru Tao
- Institute of Atherosclerosis, Taishan Institute for Hydrogen Biological Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Shucun Qin
- Institute of Atherosclerosis, Taishan Institute for Hydrogen Biological Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Xifeng Lu
- Guangdong Provincial Key Laboratory of Immunity and Diseases, Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China; Department of Physiology, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China.
| | - Qianjun He
- Guangdong Provincial Key Laboratory of Immunity and Diseases, Marshall Laboratory of Biomedical Engineering, Shenzhen University, Shenzhen, China; School of Biomedical Engineering, Health Science Center, Shenzhen University, No. 1066 Xueyuan Avenue, Shenzhen, 518060, Guangdong, China; Institute of Atherosclerosis, Taishan Institute for Hydrogen Biological Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China; Center of Hydrogen Science, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
29
|
Miller BF, Thyfault JP. Exercise-Pharmacology Interactions: Metformin, Statins, and Healthspan. Physiology (Bethesda) 2021; 35:338-347. [PMID: 32783612 DOI: 10.1152/physiol.00013.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
There is an increased focus on treatments to extend the healthspan. There is solid evidence that exercise extends the healthspan, but other treatments, such as metformin and statins, are also gaining traction. If metformin and statins will be used to prolong healthspan, we must understand their effects in those free of disease and in combination with exercise.
Collapse
Affiliation(s)
- Benjamin F Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma.,Oklahoma Nathan Shock Center for Aging, Oklahoma City, Oklahoma.,Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas.,Research Service, Kansas City VA Medical Center, Kansas City, Missouri.,Center for Children's Healthy Lifestyle and Nutrition, Children's Mercy Hospital, Kansas City, Missouri
| |
Collapse
|
30
|
The Hormetic Effect of Metformin: "Less Is More"? Int J Mol Sci 2021; 22:ijms22126297. [PMID: 34208371 PMCID: PMC8231127 DOI: 10.3390/ijms22126297] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/06/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Metformin (MTF) is the first-line therapy for type 2 diabetes (T2DM). The euglycemic effect of MTF is due to the inhibition of hepatic glucose production. Literature reports that the principal molecular mechanism of MTF is the activation of 5′-AMP-activated protein kinase (AMPK) due to the decrement of ATP intracellular content consequent to the inhibition of Complex I, although this effect is obtained only at millimolar concentrations. Conversely, micromolar MTF seems to activate the mitochondrial electron transport chain, increasing ATP production and limiting oxidative stress. This evidence sustains the idea that MTF exerts a hormetic effect based on its concentration in the target tissue. Therefore, in this review we describe the effects of MTF on T2DM on the principal target organs, such as liver, gut, adipose tissue, endothelium, heart, and skeletal muscle. In particular, data indicate that all organs, except the gut, accumulate MTF in the micromolar range when administered in therapeutic doses, unmasking molecular mechanisms that do not depend on Complex I inhibition.
Collapse
|
31
|
Khromova NV, Fedorov AV, Ma Y, Kondratov KA, Prikhodko SS, Ignatieva EV, Artemyeva MS, Anopova AD, Neimark AE, Kostareva AA, Babenko AY, Dmitrieva RI. Regulatory Action of Plasma from Patients with Obesity and Diabetes towards Muscle Cells Differentiation and Bioenergetics Revealed by the C2C12 Cell Model and MicroRNA Analysis. Biomolecules 2021; 11:769. [PMID: 34063883 PMCID: PMC8224077 DOI: 10.3390/biom11060769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are often combined and pathologically affect many tissues due to changes in circulating bioactive molecules. In this work, we evaluated the effect of blood plasma from obese (OB) patients or from obese patients comorbid with diabetes (OBD) on skeletal muscle function and metabolic state. We employed the mouse myoblasts C2C12 differentiation model to test the regulatory effect of plasma exposure at several levels: (1) cell morphology; (2) functional activity of mitochondria; (3) expression levels of several mitochondria regulators, i.e., Atgl, Pgc1b, and miR-378a-3p. Existing databases were used to computationally predict and analyze mir-378a-3p potential targets. We show that short-term exposure to OB or OBD patients' plasma is sufficient to affect C2C12 properties. In fact, the expression of genes that regulate skeletal muscle differentiation and growth was downregulated in both OB- and OBD-treated cells, maximal mitochondrial respiration rate was downregulated in the OBD group, while in the OB group, a metabolic switch to glycolysis was detected. These alterations correlated with a decrease in ATGL and Pgc1b expression in the OB group and with an increase of miR-378a-3p levels in the OBD group.
Collapse
Affiliation(s)
- Natalya V. Khromova
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Anton V. Fedorov
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Yi Ma
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Kirill A. Kondratov
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Stanislava S. Prikhodko
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Elena V. Ignatieva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Marina S. Artemyeva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Anna D. Anopova
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Aleksandr E. Neimark
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Anna A. Kostareva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
- Center for Molecular Medicine, Department of Women’s and Children’s Health, Karolinska Institute, 17177 Stockholm, Sweden
| | - Alina Yu. Babenko
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| | - Renata I. Dmitrieva
- National Almazov Medical Research Centre, Institute of Molecular Biology and Genetics, 197341 Saint-Petersburg, Russia; (A.V.F.); (Y.M.); (K.A.K.); (S.S.P.); (E.V.I.); (M.S.A.); (A.D.A.); (A.E.N.); (A.A.K.); (A.Y.B.); (R.I.D.)
| |
Collapse
|
32
|
Peralta S, Pinto M, Arguello T, Garcia S, Diaz F, Moraes CT. Metformin delays neurological symptom onset in a mouse model of neuronal complex I deficiency. JCI Insight 2020; 5:141183. [PMID: 33148885 PMCID: PMC7710273 DOI: 10.1172/jci.insight.141183] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/21/2020] [Indexed: 12/24/2022] Open
Abstract
Complex I (also known as NADH-ubiquinone oxidoreductase) deficiency is the most frequent mitochondrial disorder present in childhood. NADH-ubiquinone oxidoreductase iron-sulfur protein 3 (NDUFS3) is a catalytic subunit of the mitochondrial complex I; NDUFS3 is conserved from bacteria and essential for complex I function. Mutations affecting complex I, including in the Ndufs3 gene, cause fatal neurodegenerative diseases, such as Leigh syndrome. No treatment is available for these conditions. We developed and performed a detailed molecular characterization of a neuron-specific Ndufs3 conditional KO mouse model. We showed that deletion of Ndufs3 in forebrain neurons reduced complex I activity, altered brain energy metabolism, and increased locomotor activity with impaired motor coordination, balance, and stereotyped behavior. Metabolomics analyses showed an increase of glycolysis intermediates, suggesting an adaptive response to the complex I defect. Administration of metformin to these mice delayed the onset of the neurological symptoms but not of neuronal loss. This improvement was likely related to enhancement of glucose uptake and utilization, which are known effects of metformin in the brain. Despite reports that metformin inhibits complex I activity, our findings did not show worsening a complex I defect nor increases in lactic acid, suggesting that metformin should be further evaluated for use in patients with mitochondrial encephalopathies. Metformin delays onset of mitochondrial encephalopathy in a CNS model of mitochondrial oxidative phosphorylation defect.
Collapse
Affiliation(s)
| | | | | | | | | | - Carlos T Moraes
- Department of Neurology and.,Department of Cell Biology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
33
|
Metformin Preconditioning Improves Hepatobiliary Function and Reduces Injury in a Rat Model of Normothermic Machine Perfusion and Orthotopic Transplantation. Transplantation 2020; 104:e271-e280. [PMID: 32150043 PMCID: PMC7439933 DOI: 10.1097/tp.0000000000003216] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background. Preconditioning of donor livers before organ retrieval may improve organ quality after transplantation. We investigated whether preconditioning with metformin reduces preservation injury and improves hepatobiliary function in rat donor livers during ex situ normothermic machine perfusion (NMP) and after orthotopic liver transplantation. Methods. Lewis rats were administered metformin via oral gavage, after which a donor hepatectomy was performed followed by a standardized cold storage period of 4 hours. Graft assessment was performed using NMP via double perfusion of the hepatic artery and portal vein. In an additional experiment, rat donor livers preconditioned with metformin were stored on ice for 4 hours and transplanted to confirm postoperative liver function and survival. Data were analyzed and compared with sham-fed controls. Results. Graft assessment using NMP confirmed that preconditioning significantly improved ATP production, markers for hepatobiliary function (total bile production, biliary bilirubin, and bicarbonate), and significantly lowered levels of lactate, glucose, and apoptosis. After orthotopic liver transplantation, metformin preconditioning significantly reduced transaminase levels. Conclusions. Preconditioning with metformin lowers hepatobiliary injury and improves hepatobiliary function in an in situ and ex situ model of rat donor liver transplantation.
Collapse
|
34
|
Christian CJ, Benian GM. Animal models of sarcopenia. Aging Cell 2020; 19:e13223. [PMID: 32857472 PMCID: PMC7576270 DOI: 10.1111/acel.13223] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Sarcopenia is the age-related decline in muscle mass and function without any underlying disease. The exact molecular mechanisms responsible for this pathology remain unknown. The use of model organisms, such as mice, rats, flies, and worms, has advanced the field of sarcopenia research by identifying therapeutic strategies and genetic mutations that result in improved muscle mass and function of elderly animals. This review discusses molecular and therapeutic discoveries made using these model organisms and how these animals can be further utilized to better understand sarcopenia pathogenesis. In rodents, flies, and worms, dietary restriction improves muscle performance in old animals. In rodents and worms, exercise and a number of naturally occurring compounds alleviate sarcopenia. Reduction in the insulin/IGF1 receptor pathway, well known to promote longevity, improves sarcopenia in worms and flies. Mitochondrial dysfunction may contribute to the pathogenesis of sarcopenia: In rodents, there is age-dependent reduction in mitochondrial mass and a change in morphology; in nematodes, there is age-dependent fragmentation of mitochondria that precedes sarcomeric disorganization. In Drosophila and rats, components of the 26S proteasome are elevated in aged muscle. An advantage of the worm and fly models is that these organisms lack muscle stem cells, and thus processes that promote the maintenance of already assembled muscle, can be identified without the confounding influence of muscle regeneration. Zebrafish are an up and coming model of sarcopenia for future consideration. A better understanding of the molecular changes behind sarcopenia will help researchers develop better therapies to improve the muscle health of elderly individuals.
Collapse
Affiliation(s)
| | - Guy M. Benian
- Department of Pathology Emory University Atlanta Georgia USA
| |
Collapse
|
35
|
Ryan TE, Schmidt CA, Tarpey MD, Amorese AJ, Yamaguchi DJ, Goldberg EJ, Iñigo MM, Karnekar R, O'Rourke A, Ervasti JM, Brophy P, Green TD, Neufer PD, Fisher-Wellman K, Spangenburg EE, McClung JM. PFKFB3-mediated glycolysis rescues myopathic outcomes in the ischemic limb. JCI Insight 2020; 5:139628. [PMID: 32841216 PMCID: PMC7526546 DOI: 10.1172/jci.insight.139628] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/19/2020] [Indexed: 12/24/2022] Open
Abstract
Compromised muscle mitochondrial metabolism is a hallmark of peripheral arterial disease, especially in patients with the most severe clinical manifestation - critical limb ischemia (CLI). We asked whether inflexibility in metabolism is critical for the development of myopathy in ischemic limb muscles. Using Polg mtDNA mutator (D257A) mice, we reveal remarkable protection from hind limb ischemia (HLI) due to a unique and beneficial adaptive enhancement of glycolytic metabolism and elevated ischemic muscle PFKFB3. Similar to the relationship between mitochondria from CLI and claudicating patient muscles, BALB/c muscle mitochondria are uniquely dysfunctional after HLI onset as compared with the C57BL/6 (BL6) parental strain. AAV-mediated overexpression of PFKFB3 in BALB/c limb muscles improved muscle contractile function and limb blood flow following HLI. Enrichment analysis of RNA sequencing data on muscle from CLI patients revealed a unique deficit in the glucose metabolism Reactome. Muscles from these patients express lower PFKFB3 protein, and their muscle progenitor cells possess decreased glycolytic flux capacity in vitro. Here, we show supplementary glycolytic flux as sufficient to protect against ischemic myopathy in instances where reduced blood flow-related mitochondrial function is compromised preclinically. Additionally, our data reveal reduced glycolytic flux as a common characteristic of the failing CLI patient limb skeletal muscle.
Collapse
Affiliation(s)
- Terence E Ryan
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - Cameron A Schmidt
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - Michael D Tarpey
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - Adam J Amorese
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - Dean J Yamaguchi
- Department of Cardiovascular Science, and.,Division of Surgery, East Carolina University, Brody School of Medicine, Greenville, North Carolina, USA
| | - Emma J Goldberg
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - Melissa Mr Iñigo
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - Reema Karnekar
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - Allison O'Rourke
- Department of Biochemistry, Molecular Biology and Biophysics, College of Biological Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, College of Biological Sciences, University of Minnesota, Saint Paul, Minnesota, USA
| | | | - Thomas D Green
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute.,Department of Physiology
| | | | | | - Joseph M McClung
- East Carolina Diabetes and Obesity Institute.,Department of Physiology.,Department of Cardiovascular Science, and
| |
Collapse
|
36
|
p38 MAPK in Glucose Metabolism of Skeletal Muscle: Beneficial or Harmful? Int J Mol Sci 2020; 21:ijms21186480. [PMID: 32899870 PMCID: PMC7555282 DOI: 10.3390/ijms21186480] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/27/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscles respond to environmental and physiological changes by varying their size, fiber type, and metabolic properties. P38 mitogen-activated protein kinase (MAPK) is one of several signaling pathways that drive the metabolic adaptation of skeletal muscle to exercise. p38 MAPK also participates in the development of pathological traits resulting from excessive caloric intake and obesity that cause metabolic syndrome and type 2 diabetes (T2D). Whereas p38 MAPK increases insulin-independent glucose uptake and oxidative metabolism in muscles during exercise, it contrastingly mediates insulin resistance and glucose intolerance during metabolic syndrome development. This article provides an overview of the apparent contradicting roles of p38 MAPK in the adaptation of skeletal muscles to exercise and to pathological conditions leading to glucose intolerance and T2D. Here, we focus on the involvement of p38 MAPK in glucose metabolism of skeletal muscle, and discuss the possibility of targeting this pathway to prevent the development of T2D.
Collapse
|
37
|
Mitochondrial dysfunction in the fetoplacental unit in gestational diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165948. [PMID: 32866635 DOI: 10.1016/j.bbadis.2020.165948] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Gestational diabetes mellitus (GDM) is a disease of pregnancy that is associated with d-glucose intolerance and foeto-placental vascular dysfunction. GMD causes mitochondrial dysfunction in the placental endothelium and trophoblast. Additionally, GDM is associated with reduced placental oxidative phosphorylation due to diminished activity of the mitochondrial F0F1-ATP synthase (complex V). This phenomenon may result from a higher generation of reactive superoxide anion and nitric oxide. Placental mitochondrial biogenesis and mitophagy work in concert to maintain cell homeostasis and are vital mechanisms securing the efficient generation of ATP, whose demand is higher in pregnancy, ensuring foetal growth and development. Additional factors disturbing placental ATP synthase activity in GDM include pre-gestational maternal obesity or overweight, intracellular pH, miRNAs, fatty acid oxidation, and foetal (and 'placental') sex. GDM is also associated with maternal and foetal hyperinsulinaemia, altered circulating levels of adiponectin and leptin, and the accumulation of extracellular adenosine. Here, we reviewed the potential interplay between these molecules or metabolic conditions on the mechanisms of mitochondrial dysfunction in the foeto-placental unit in GDM pregnancies.
Collapse
|
38
|
Wang L, Halliday G, Huot JR, Satoh T, Baust JJ, Fisher A, Cook T, Hu J, Avolio T, Goncharov DA, Bai Y, Vanderpool RR, Considine RV, Bonetto A, Tan J, Bachman TN, Sebastiani A, Mora AL, Machado RF, Goncharova EA, Gladwin MT, Lai YC. Treatment With Treprostinil and Metformin Normalizes Hyperglycemia and Improves Cardiac Function in Pulmonary Hypertension Associated With Heart Failure With Preserved Ejection Fraction. Arterioscler Thromb Vasc Biol 2020; 40:1543-1558. [PMID: 32268788 PMCID: PMC7255946 DOI: 10.1161/atvbaha.119.313883] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Pulmonary hypertension (PH) due to left heart disease (group 2), especially in the setting of heart failure with preserved ejection fraction (HFpEF), is the most common cause of PH worldwide; however, at present, there is no proven effective therapy available for its treatment. PH-HFpEF is associated with insulin resistance and features of metabolic syndrome. The stable prostacyclin analog, treprostinil, is an effective and widely used Food and Drug Administration-approved drug for the treatment of pulmonary arterial hypertension. While the effect of treprostinil on metabolic syndrome is unknown, a recent study suggests that the prostacyclin analog beraprost can improve glucose intolerance and insulin sensitivity. We sought to evaluate the effectiveness of treprostinil in the treatment of metabolic syndrome-associated PH-HFpEF. Approach and Results: Treprostinil treatment was given to mice with mild metabolic syndrome-associated PH-HFpEF induced by high-fat diet and to SU5416/obese ZSF1 rats, a model created by the treatment of rats with a more profound metabolic syndrome due to double leptin receptor defect (obese ZSF1) with a vascular endothelial growth factor receptor blocker SU5416. In high-fat diet-exposed mice, chronic treatment with treprostinil reduced hyperglycemia and pulmonary hypertension. In SU5416/Obese ZSF1 rats, treprostinil improved hyperglycemia with similar efficacy to that of metformin (a first-line drug for type 2 diabetes mellitus); the glucose-lowering effect of treprostinil was further potentiated by the combined treatment with metformin. Early treatment with treprostinil in SU5416/Obese ZSF1 rats lowered pulmonary pressures, and a late treatment with treprostinil together with metformin improved pulmonary artery acceleration time to ejection time ratio and tricuspid annular plane systolic excursion with AMPK (AMP-activated protein kinase) activation in skeletal muscle and the right ventricle. CONCLUSIONS Our data suggest a potential use of treprostinil as an early treatment for mild metabolic syndrome-associated PH-HFpEF and that combined treatment with treprostinil and metformin may improve hyperglycemia and cardiac function in a more severe disease.
Collapse
Affiliation(s)
- Longfei Wang
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
- The Third Xiangya Hospital, Central South University; Changsha, Hunan, China
| | - Gunner Halliday
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine
| | - Joshua R. Huot
- Department of Surgery, Indiana University School of Medicine
| | - Taijyu Satoh
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jeff J. Baust
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
| | - Amanda Fisher
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine
| | - Todd Cook
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine
| | - Jian Hu
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
| | - Theodore Avolio
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
| | - Dmitry A. Goncharov
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
| | - Yang Bai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine
- Department of Clinical Pharmacology, College of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | | | | | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine
| | - Jiangning Tan
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh
| | - Timothy N. Bachman
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
| | - Andrea Sebastiani
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
| | - Ana L. Mora
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine
| | - Elena A. Goncharova
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh
| | - Mark T. Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Indiana University School of Medicine
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine
| |
Collapse
|
39
|
Ramos JS, Dalleck LC, Keith CE, Fennell M, Lee Z, Drummond C, Keating SE, Fassett RG, Coombes JS. Optimizing the Interaction of Exercise Volume and Metformin to Induce a Clinically Significant Reduction in Metabolic Syndrome Severity: A Randomised Trial. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17103695. [PMID: 32456272 PMCID: PMC7277162 DOI: 10.3390/ijerph17103695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/11/2020] [Accepted: 05/22/2020] [Indexed: 12/20/2022]
Abstract
Insulin resistance is a central mediating factor of the metabolic syndrome (MetS), with exercise training and metformin proven antidotes to insulin resistance. However, when the two therapies are combined there is conflicting data regarding whether metformin blunts or improves exercise training-induced adaptations. The volume of exercise (duration, intensity, and frequency) on the interaction of exercise training and metformin has yet to be investigated. The aim of this study is therefore to explore the impact of a combination of different exercise volumes and metformin on MetS severity. This is a secondary analysis of data from one of the sites of the ‘Exercise in Prevention of Metabolic Syndrome’ (EX-MET) study. Ninety-nine adults with MetS were randomized into a 16-week exercise program completing either: (i) moderate-intensity continuous training (MICT) at 60–70% of peak heart rate (HRpeak) for 30 min/session (n = 34, 150 min/week); (ii) high-volume high-intensity interval training (HIIT) consisting of 4 × 4 min bouts at 85–95% HRpeak, interspersed with 3 min of active recovery at 50–70% HRpeak (n = 34, 38 min/session, 114 min/week); or (iii) low volume HIIT, 1 × 4 min bout of HIIT at 85–95% HRpeak (n = 31, 17 min/session, 51 min/week). Metformin intake was monitored and recorded throughout the trial. MetS severity was calculated as z-scores derived from MetS risk factors assessed at pre- and post-intervention. Sixty-five participants had complete pre- and post-intervention data for MetS z-score, of which 18 participants (28%) were taking metformin. Over the 16-week intervention, a similar proportion of participants clinically improved MetS severity (Δ ≥ −0.87) with metformin (8/18, 44%) or without metformin (23/47, 49%) (p = 0.75). While there were no between-group differences (p = 0.24), in those who did not take metformin low-volume HIIT had more likely responders (10/15, 67%) compared to MICT (6/16, 38%) and high-volume HIIT (7/16, 44%). In those taking metformin, there was a lower proportion of participants who clinically improved MetS severity following high-volume HIIT (1/6, 17%) compared to MICT (2/4, 50%) and low-volume HIIT (5/8, 63%), but with no between-group difference (p = 0.23). Moreover, in those who performed high-volume HIIT, there was a statistically significantly higher proportion (p = 0.03) of likely non-responders with improved MetS severity in participants taking metformin (4/6, 67%) compared to those not taking metformin (3/16, 19%). In individuals with MetS, the effect of high volume HIIT on MetS severity may be blunted in those taking metformin. These findings need to be confirmed in a larger study.
Collapse
Affiliation(s)
- Joyce S. Ramos
- Caring Futures Institute & SHAPE Research Centre, Exercise Science and Clinical Exercise Physiology, College of Nursing and Health Sciences, Flinders University, Room S268, South Wing, Sturt Building, Bedford Park, Adelaide 5042, South Australia, Australia; (L.C.D.); (C.E.K.); (M.F.); (Z.L.); (C.D.)
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Queensland, Australia; (S.E.K.); (R.G.F.); (J.S.C.)
- Correspondence: ; Tel.: +61-435-942-505
| | - Lance C. Dalleck
- Caring Futures Institute & SHAPE Research Centre, Exercise Science and Clinical Exercise Physiology, College of Nursing and Health Sciences, Flinders University, Room S268, South Wing, Sturt Building, Bedford Park, Adelaide 5042, South Australia, Australia; (L.C.D.); (C.E.K.); (M.F.); (Z.L.); (C.D.)
- Recreation, Exercise, and Sport Science Department, Western Colorado University, Gunnison, CO 81231, USA
| | - Caitlin E. Keith
- Caring Futures Institute & SHAPE Research Centre, Exercise Science and Clinical Exercise Physiology, College of Nursing and Health Sciences, Flinders University, Room S268, South Wing, Sturt Building, Bedford Park, Adelaide 5042, South Australia, Australia; (L.C.D.); (C.E.K.); (M.F.); (Z.L.); (C.D.)
| | - Mackenzie Fennell
- Caring Futures Institute & SHAPE Research Centre, Exercise Science and Clinical Exercise Physiology, College of Nursing and Health Sciences, Flinders University, Room S268, South Wing, Sturt Building, Bedford Park, Adelaide 5042, South Australia, Australia; (L.C.D.); (C.E.K.); (M.F.); (Z.L.); (C.D.)
| | - Zoe Lee
- Caring Futures Institute & SHAPE Research Centre, Exercise Science and Clinical Exercise Physiology, College of Nursing and Health Sciences, Flinders University, Room S268, South Wing, Sturt Building, Bedford Park, Adelaide 5042, South Australia, Australia; (L.C.D.); (C.E.K.); (M.F.); (Z.L.); (C.D.)
| | - Claire Drummond
- Caring Futures Institute & SHAPE Research Centre, Exercise Science and Clinical Exercise Physiology, College of Nursing and Health Sciences, Flinders University, Room S268, South Wing, Sturt Building, Bedford Park, Adelaide 5042, South Australia, Australia; (L.C.D.); (C.E.K.); (M.F.); (Z.L.); (C.D.)
| | - Shelley E. Keating
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Queensland, Australia; (S.E.K.); (R.G.F.); (J.S.C.)
| | - Robert G. Fassett
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Queensland, Australia; (S.E.K.); (R.G.F.); (J.S.C.)
| | - Jeff S. Coombes
- Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane 4072, Queensland, Australia; (S.E.K.); (R.G.F.); (J.S.C.)
| |
Collapse
|
40
|
Metformin: A Possible Option in Cancer Chemotherapy. Anal Cell Pathol (Amst) 2020; 2020:7180923. [PMID: 32399389 PMCID: PMC7201450 DOI: 10.1155/2020/7180923] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/20/2020] [Accepted: 04/10/2020] [Indexed: 02/07/2023] Open
Abstract
Metformin has been used for a long time as an antidiabetic medication for type 2 diabetes. It is used either as a monotherapy or in combination with other antidiabetic medications. The drug came into prominence in diabetes and other conditions with cardiovascular risk after the landmark study of 1995 by the United Kingdom Prospective Diabetes Study which emphasized its importance. However, the drug has been used in experimental trials in various aspects of medicine and pharmacology such as in reproductive medicine, cancer chemotherapy, metabolic diseases, and neurodegenerative diseases. It has been in use in the treatment of polycystic ovarian disease and obesity and is being considered in type 1 diabetes. This study seeks to evaluate the relevance of metformin in cancer management. Different mechanisms have been proposed for its antitumor action which involves the following: (a) the activation of adenosine monophosphate kinase, (b) modulation of adenosine A1 receptor (ADORA), (c) reduction in insulin/insulin growth factors, and (d) the role of metformin in the inhibition of endogenous reactive oxygen species (ROS); and its resultant damage to deoxyribonucleic acid (DNA) molecule is another paramount antitumor mechanism.
Collapse
|
41
|
Chung E, Elmassry MM, Kottapalli P, Kottapalli KR, Kaur G, Dufour JM, Wright K, Ramalingam L, Moustaid-Moussa N, Wang R, Hamood AN, Shen CL. Metabolic benefits of annatto-extracted tocotrienol on glucose homeostasis, inflammation, and gut microbiome. Nutr Res 2020; 77:97-107. [PMID: 32438021 DOI: 10.1016/j.nutres.2020.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/27/2020] [Accepted: 04/11/2020] [Indexed: 12/12/2022]
Abstract
Emerging evidence suggests that the gut microbiome plays an important role in the pathophysiology of both obesity and type 2 diabetes mellitus. We previously reported that dietary annatto-extracted tocotrienol exerts beneficial effects by modulating inflammatory responses in mice fed a high-fat diet (HFD). The purpose of this study was to test the hypothesis that tocotrienol supplementation when combined with an HFD would result in an altered gut microbiota composition. For 14 weeks, forty-eight male C57BL/6J mice were assigned to 4 groups-low-fat diet, HFD, HFD supplemented with annatto-extracted tocotrienol at 800 mg/kg diet (AT), and HFD supplemented with metformin at 200 mg/kg diet. Glucose homeostasis was assessed by glucose and insulin tolerance tests, serum and pancreas insulin levels, and histological assessments of insulin and glucagon in pancreatic tissue. The concentrations of adipokines were measured in white adipose tissues. For the gut microbiome analysis, cecal content was collected, DNA was extracted, and 16S rRNA gene sequencing was performed. AT supplementation improved glucose homeostasis and lowered resistin, leptin, and interleukin-6 levels in white adipose tissue. Relative to the HFD group, AT-supplemented mice showed a decrease in the Firmicutes to Bacteroidetes ratio and had a lower abundance of Ruminococcus lactaris, Dorea longicatena, and Lachnospiraceae family. The relative abundance of Akkermansia muciniphila was increased in the AT group compared to the low-fat diet group. The association between the metabolic improvements and the identified bacterial taxa suggests a potential metabolic modulation caused by AT supplementation through the gut microbiota composition in mice fed an HFD.
Collapse
Affiliation(s)
- Eunhee Chung
- Department of Kinesiology, Health, and Nutrition, University of Texas at San Antonio, San Antonio, TX.
| | - Moamen M Elmassry
- Department of Biological Sciences, Texas Tech University, Lubbock, TX.
| | | | | | - Gurvinder Kaur
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX; Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX.
| | - Jannette M Dufour
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX; Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Kandis Wright
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Latha Ramalingam
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Nutritional Sciences, Texas Tech University, Lubbock, TX.
| | - Naima Moustaid-Moussa
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Nutritional Sciences, Texas Tech University, Lubbock, TX.
| | - Rui Wang
- Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Abdul N Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX; Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX.
| | - Chwan-Li Shen
- Center of Excellence for Integrative Health, Texas Tech University Health Sciences Center, Lubbock, TX; Obesity Research Institute, Texas Tech University, Lubbock, TX; Department of Pathology, Texas Tech University Health Sciences Center, Lubbock, TX.
| |
Collapse
|
42
|
Abstract
Doxorubicin is a commonly used chemotherapeutic agent for the treatment of a range of cancers, but despite its success in improving cancer survival rates, doxorubicin is cardiotoxic and can lead to congestive heart failure. Therapeutic options for this patient group are limited to standard heart failure medications with the only drug specific for doxorubicin cardiotoxicity to reach FDA approval being dexrazoxane, an iron-chelating agent targeting oxidative stress. However, dexrazoxane has failed to live up to its expectations from preclinical studies while also bringing up concerns about its safety. Despite decades of research, the molecular mechanisms of doxorubicin cardiotoxicity are still poorly understood and oxidative stress is no longer considered to be the sole evil. Mitochondrial impairment, increased apoptosis, dysregulated autophagy and increased fibrosis have also been shown to be crucial players in doxorubicin cardiotoxicity. These cellular processes are all linked by one highly conserved intracellular kinase: adenosine monophosphate-activated protein kinase (AMPK). AMPK regulates mitochondrial biogenesis via PGC1α signalling, increases oxidative mitochondrial metabolism, decreases apoptosis through inhibition of mTOR signalling, increases autophagy through ULK1 and decreases fibrosis through inhibition of TGFβ signalling. AMPK therefore sits at the control point of many mechanisms shown to be involved in doxorubicin cardiotoxicity and cardiac AMPK signalling itself has been shown to be impaired by doxorubicin. In this review, we introduce different agents known to activate AMPK (metformin, statins, resveratrol, thiazolidinediones, AICAR, specific AMPK activators) as well as exercise and dietary restriction, and we discuss the existing evidence for their potential role in cardioprotection from doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Kerstin N Timm
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK.
| | - Damian J Tyler
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| |
Collapse
|
43
|
Xie J, Ye J, Cai Z, Luo Y, Zhu X, Deng Y, Feng Y, Liang Y, Liu R, Han Z, Liang Y, Zheng Y, Mo R, Zhuo Y, Wu Y, Jiang F, Zhu J, Wu CL, Zhong W. GPD1 Enhances the Anticancer Effects of Metformin by Synergistically Increasing Total Cellular Glycerol-3-Phosphate. Cancer Res 2020; 80:2150-2162. [PMID: 32179514 DOI: 10.1158/0008-5472.can-19-2852] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/06/2020] [Accepted: 03/11/2020] [Indexed: 11/16/2022]
Abstract
Metformin is an oral drug widely used for the treatment of type 2 diabetes mellitus. Numerous studies have demonstrated the value of metformin in cancer treatment. However, for metformin to elicit effects on cancer often requires a high dosage, and any underlying mechanism for how to improve its inhibitory effects remains unknown. Here, we found that low mRNA expression of glycerol-3-phosphate dehydrogenase 1 (GPD1) may predict a poor response to metformin treatment in 15 cancer cell lines. In vitro and in vivo, metformin treatment alone significantly suppressed cancer cell proliferation, a phenotype enhanced by GPD1 overexpression. Total cellular glycerol-3-phosphate concentration was significantly increased by the combination of GPD1 overexpression and metformin treatment, which suppressed cancer growth via inhibition of mitochondrial function. Eventually, increased reactive oxygen species and mitochondrial structural damage was observed in GPD1-overexpressing cell lines treated with metformin, which may contribute to cell death. In summary, this study demonstrates that GPD1 overexpression enhances the anticancer activity of metformin and that patients with increased GPD1 expression in tumor cells may respond better to metformin therapy. SIGNIFICANCE: GPD1 overexpression enhances the anticancer effect of metformin through synergistic inhibition of mitochondrial function, thereby providing new insight into metformin-mediated cancer therapy.
Collapse
Affiliation(s)
- Jianjiang Xie
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zhiduan Cai
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Luo
- Department of Urology, the Second People's Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Xuejin Zhu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yulin Deng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yingke Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ren Liu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhaodong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuxiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yu Zheng
- Department of Urology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Rujun Mo
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yangjia Zhuo
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yongding Wu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Funeng Jiang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianguo Zhu
- Department of Urology, Guizhou Provincial People's Hospital, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
- School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
44
|
Chen F, Xu S, Wang Y, Chen F, Cao L, Liu T, Huang T, Wei Q, Ma G, Zhao Y, Wang D. Risk Factors for Sarcopenia in the Elderly with Type 2 Diabetes Mellitus and the Effect of Metformin. J Diabetes Res 2020; 2020:3950404. [PMID: 33083494 PMCID: PMC7563046 DOI: 10.1155/2020/3950404] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/23/2020] [Indexed: 12/19/2022] Open
Abstract
AIMS Sarcopenia is a common condition in older individuals, especially in the elderly with type 2 diabetes mellitus (T2DM). The aim of the present study was to examine the risk factors for sarcopenia in elderly individuals with T2DM and the effects of metformin. METHODS A total of 1732 elderly with T2DM were recruited to this cross-sectional observational study, and we analyzed the data using logistic regression analyses. Skeletal muscle mass, grip strength, and usual gait speed were measured to diagnose sarcopenia according to the criteria of the Asian Working Group for Sarcopenia, combined with expert consensus on sarcopenia in China. RESULTS The overall prevalence of sarcopenia was 10.37% of the participants. In the multivariate analysis, sex, age, educational level, and BMI were risk factors for sarcopenia, with women more likely to develop sarcopenia relative to men (OR = 2.539, 95% CI = 1.475-4.371; P < 0.05). We observed that sarcopenia increased with age and decreased with increasing BMI and educational level (P < 0.05). Participants who took metformin alone or combined with other drugs exhibited a lower risk for sarcopenia than those who took no medication (OR = 0.510, 95% CI = 0.288-0.904 and OR = 0.398, 95% CI = 0.225-0.702, respectively; P < 0.05). CONCLUSIONS We showed that being female and at an older age, lower educational level, and lower BMI were risk factors for sarcopenia in elderly T2DM and that metformin acted as a protective agent against sarcopenia in these patients.
Collapse
Affiliation(s)
- Fenqin Chen
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Shuai Xu
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Yingfang Wang
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Feng Chen
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Lu Cao
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Tingting Liu
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Ting Huang
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Qian Wei
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Guojing Ma
- Departments of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang 110001, China
| | - Yuhong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- China Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Difei Wang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
45
|
Anoveros‐Barrera A, Bhullar AS, Stretch C, Esfandiari N, Dunichand‐Hoedl AR, Martins KJ, Bigam D, Khadaroo RG, McMullen T, Bathe OF, Damaraju S, Skipworth RJ, Putman CT, Baracos VE, Mazurak VC. Clinical and biological characterization of skeletal muscle tissue biopsies of surgical cancer patients. J Cachexia Sarcopenia Muscle 2019; 10:1356-1377. [PMID: 31307124 PMCID: PMC9536086 DOI: 10.1002/jcsm.12466] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/10/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Researchers increasingly use intraoperative muscle biopsy to investigate mechanisms of skeletal muscle atrophy in patients with cancer. Muscles have been assessed for morphological, cellular, and biochemical features. The aim of this study was to conduct a state-of-the-science review of this literature and, secondly, to evaluate clinical and biological variation in biopsies of rectus abdominis (RA) muscle from a cohort of patients with malignancies. METHODS Literature was searched for reports on muscle biopsies from patients with a cancer diagnosis. Quality of reports and risk of bias were assessed. Data abstracted included patient characteristics and diagnoses, sample size, tissue collection and biobanking procedures, and results. A cohort of cancer patients (n = 190, 88% gastrointestinal malignancies), who underwent open abdominal surgery as part of their clinical care, consented to RA biopsy from the site of incision. Computed tomography (CT) scans were used to quantify total abdominal muscle and RA cross-sectional areas and radiodensity. Biopsies were assessed for muscle fibre area (μm2 ), fibre types, myosin heavy chain isoforms, and expression of genes selected for their involvement in catabolic pathways of muscle. RESULTS Muscle biopsy occurred in 59 studies (total N = 1585 participants). RA was biopsied intraoperatively in 40 studies (67%), followed by quadriceps (26%; percutaneous biopsy) and other muscles (7%). Cancer site and stage, % of male participants, and age were highly variable between studies. Details regarding patient medical history and biopsy procedures were frequently absent. Lack of description of the population(s) sampled and low sample size contributed to low quality and risk of bias. Weight-losing cases were compared with weight stable cancer or healthy controls without considering a measure of muscle mass in 21 out of 44 studies. In the cohort of patients providing biopsy for this study, 78% of patients had preoperative CT scans and a high proportion (64%) met published criteria for sarcopenia. Fibre type distribution in RA was type I (46% ± 13), hybrid type I/IIA (1% ± 1), type IIA (36% ± 10), hybrid type IIA/D (15% ± 14), and type IID (2% ± 5). Sexual dimorphism was prominent in RA CT cross-sectional area, mean fibre cross-sectional area, and in expression of genes associated with muscle growth, apoptosis, and inflammation (P < 0.05). Medical history revealed multiple co-morbid conditions and medications. CONCLUSIONS Continued collaboration between researchers and cancer surgeons enables a more complete understanding of mechanisms of cancer-associated muscle atrophy. Standardization of biobanking practices, tissue manipulation, patient characterization, and classification will enhance the consistency, reliability, and comparability of future studies.
Collapse
Affiliation(s)
- Ana Anoveros‐Barrera
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | - Amritpal S. Bhullar
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | | | - Nina Esfandiari
- Department of Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Abha R. Dunichand‐Hoedl
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | - Karen J.B. Martins
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| | - David Bigam
- Department of Surgery, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Rachel G. Khadaroo
- Department of Surgery, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Todd McMullen
- Department of Surgery, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Oliver F. Bathe
- Department of OncologyUniversity of CalgaryCalgaryABCanada
- Department of SurgeryUniversity of CalgaryCalgaryABCanada
| | - Sambasivarao Damaraju
- Department of Laboratory Medicine and PathologyUniversity of AlbertaEdmontonABCanada
- Department of Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | | | - Charles T. Putman
- Faculty of Kinesiology, Sport, and Recreation, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Vickie E. Baracos
- Department of Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonABCanada
| | - Vera C. Mazurak
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental SciencesUniversity of AlbertaEdmontonABCanada
| |
Collapse
|
46
|
Hernández-Álvarez D, Mena-Montes B, Toledo-Pérez R, Pedraza-Vázquez G, López-Cervantes SP, Morales-Salazar A, Hernández-Cruz E, Lazzarini-Lechuga R, Vázquez-Cárdenas RR, Vilchis-DeLaRosa S, Posadas-Rodríguez P, Santín-Márquez R, Rosas-Carrasco O, Ibañez-Contreras A, Alarcón-Aguilar A, López-Díazguerrero NE, Luna-López A, Königsberg M. Long-Term Moderate Exercise Combined with Metformin Treatment Induces an Hormetic Response That Prevents Strength and Muscle Mass Loss in Old Female Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3428543. [PMID: 31814870 PMCID: PMC6877950 DOI: 10.1155/2019/3428543] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/18/2019] [Indexed: 12/18/2022]
Abstract
Sarcopenia is a syndrome characterized by a progressive and generalized skeletal muscle mass and strength loss, as well as a poor physical performance, which as strongly been associated with aging. Sedentary lifestyle in the elderly contributes to this condition; however, physical activity improves health, reducing morbidity and mortality. Recent studies have shown that metformin (MTF) can also prevent muscle damage promoting muscular performance. To date, there is great controversy if MTF treatment combined with exercise training improves or nullifies the benefits provided by physical activity. This study is aimed at evaluating the effect of long-term moderate exercise combined with MTF treatment on body composition, strength, redox state, and survival rate during the life of female Wistar rats. In this study, rats performed moderate exercise during 20 of their 24 months of life and were treated with MTF for one year or for 6 months, i.e., from 12 to 24 months old and 18 to 24 months old. The body composition (percentage of fat, bone, and lean mass) was determined using a dual-energy X-ray absorption scanner (DXA), and grip strength was determined using a dynamometer. Likewise, medial and tibial nerve somatosensory evoked potentials were evaluated and the redox state was measured by HPLC, calculating the GSH/GSSG ratio in the gastrocnemius muscle. Our results suggest- that the MTF administration, both in the sedentary and the exercise groups, might activate a mechanism that is directly related to the induction of the hormetic response through the redox state modulation. MTF treatment does not eliminate the beneficial effects of exercise throughout life, and although MTF does not increase muscle mass, it increases longevity.
Collapse
Affiliation(s)
- David Hernández-Álvarez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Posgrado en Ciencias Biológicas, Universidad Autónoma Metropolitana, Ciudad de México 09340, Mexico
| | - Beatriz Mena-Montes
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Instituto Nacional de Geriatría, SSA, Ciudad de México 10200, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Rafael Toledo-Pérez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Gibrán Pedraza-Vázquez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Stefanie Paola López-Cervantes
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Alfredo Morales-Salazar
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Edith Hernández-Cruz
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Roberto Lazzarini-Lechuga
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Roman Royer Vázquez-Cárdenas
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Silvia Vilchis-DeLaRosa
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Pedro Posadas-Rodríguez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | - Roberto Santín-Márquez
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | | | | | - Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| | | | | | - Mina Königsberg
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México 09340, Mexico
| |
Collapse
|
47
|
VERBOVEN KENNETH, WENS INEZ, VANDENABEELE FRANK, STEVENS AN, CELIE BERT, LAPAUW BRUNO, DENDALE PAUL, VAN LOON LUCJC, CALDERS PATRICK, HANSEN DOMINIQUE. Impact of Exercise–Nutritional State Interactions in Patients with Type 2 Diabetes. Med Sci Sports Exerc 2019; 52:720-728. [DOI: 10.1249/mss.0000000000002165] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Lewis MT, Kasper JD, Bazil JN, Frisbee JC, Wiseman RW. Quantification of Mitochondrial Oxidative Phosphorylation in Metabolic Disease: Application to Type 2 Diabetes. Int J Mol Sci 2019; 20:E5271. [PMID: 31652915 PMCID: PMC6862501 DOI: 10.3390/ijms20215271] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/17/2019] [Accepted: 10/22/2019] [Indexed: 12/17/2022] Open
Abstract
Type 2 diabetes (T2D) is a growing health concern with nearly 400 million affected worldwide as of 2014. T2D presents with hyperglycemia and insulin resistance resulting in increased risk for blindness, renal failure, nerve damage, and premature death. Skeletal muscle is a major site for insulin resistance and is responsible for up to 80% of glucose uptake during euglycemic hyperglycemic clamps. Glucose uptake in skeletal muscle is driven by mitochondrial oxidative phosphorylation and for this reason mitochondrial dysfunction has been implicated in T2D. In this review we integrate mitochondrial function with physiologic function to present a broader understanding of mitochondrial functional status in T2D utilizing studies from both human and rodent models. Quantification of mitochondrial function is explained both in vitro and in vivo highlighting the use of proper controls and the complications imposed by obesity and sedentary lifestyle. This review suggests that skeletal muscle mitochondria are not necessarily dysfunctional but limited oxygen supply to working muscle creates this misperception. Finally, we propose changes in experimental design to address this question unequivocally. If mitochondrial function is not impaired it suggests that therapeutic interventions and drug development must move away from the organelle and toward the cardiovascular system.
Collapse
Affiliation(s)
- Matthew T Lewis
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| | - Jonathan D Kasper
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Present address: Molecular Physiology Institute, Duke University, Durham, NC 27701, USA.
| | - Jason N Bazil
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
| | - Jefferson C Frisbee
- Department of Medical Biophysics, University of Western Ontario, London, ON N6A 3K7, Canada.
| | - Robert W Wiseman
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
49
|
Pino MF, Stephens NA, Eroshkin AM, Yi F, Hodges A, Cornnell HH, Pratley RE, Smith SR, Wang M, Han X, Coen PM, Goodpaster BH, Sparks LM. Endurance training remodels skeletal muscle phospholipid composition and increases intrinsic mitochondrial respiration in men with Type 2 diabetes. Physiol Genomics 2019; 51:586-595. [PMID: 31588872 DOI: 10.1152/physiolgenomics.00014.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The effects of exercise training on the skeletal muscle (SKM) lipidome and mitochondrial function have not been thoroughly explored in individuals with Type 2 diabetes (T2D). We hypothesize that 10 wk of supervised endurance training improves SKM mitochondrial function and insulin sensitivity that are related to alterations in lipid signatures within SKM of T2D (males n = 8). We employed integrated multi-omics data analyses including ex vivo lipidomics (MS/MS-shotgun) and transcriptomics (RNA-Seq). From biopsies of SKM, tissue and primary myotubes mitochondrial respiration were quantified by high-resolution respirometry. We also performed hyperinsulinemic-euglycemic clamps and blood draws before and after the training. The lipidomics analysis revealed that endurance training (>95% compliance) increased monolysocardiolipin by 68.2% (P ≤ 0.03), a putative marker of mitochondrial remodeling, and reduced total sphingomyelin by 44.8% (P ≤ 0.05) and phosphatidylserine by 39.7% (P ≤ 0.04) and tended to reduce ceramide lipid content by 19.8%. Endurance training also improved intrinsic mitochondrial respiration in SKM of T2D without alterations in mitochondrial DNA copy number or cardiolipin content. RNA-Seq revealed 71 transcripts in SKM of T2D that were differentially regulated. Insulin sensitivity was unaffected, and HbA1c levels moderately increased by 7.3% despite an improvement in cardiorespiratory fitness (V̇o2peak) following the training intervention. In summary, endurance training improves intrinsic and cell-autonomous SKM mitochondrial function and modifies lipid composition in men with T2D independently of alterations in insulin sensitivity and glycemic control.
Collapse
Affiliation(s)
- Maria F Pino
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Natalie A Stephens
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Alexey M Eroshkin
- Sanford Burnham Prebys Medical Discovery Institute, Torrey Pines, California
| | - Fanchao Yi
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Andrew Hodges
- Sanford Burnham Prebys Medical Discovery Institute, Torrey Pines, California
| | - Heather H Cornnell
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Miao Wang
- University of Texas Health Sciences Center San Antonio, San Antonio, Texas
| | - Xianlin Han
- University of Texas Health Sciences Center San Antonio, San Antonio, Texas
| | - Paul M Coen
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Bret H Goodpaster
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| | - Lauren M Sparks
- Translational Research Institute for Metabolism and Diabetes, Adventhealth, Orlando, Florida
| |
Collapse
|
50
|
Stefanaki C, Bacopoulou F, Kandaraki E, Boschiero D, Diamandi-Kandarakis E. Lean Women on Metformin and Oral Contraceptives for Polycystic Ovary Syndrome Demonstrate a Dehydrated Osteosarcopenic Phenotype: A Pilot Study. Nutrients 2019; 11:nu11092055. [PMID: 31480705 PMCID: PMC6769734 DOI: 10.3390/nu11092055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/25/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022] Open
Abstract
Scarce data exist on the body composition of lean women with polycystic ovary syndrome (PCOS) on treatment with metformin and oral contraceptives (OCs). Thirty-four lean (body mass index 18.5–24.9 kg/m2) women (17 with PCOS on metformin and OCs treatment for six months and 17 controls) aged 18–40 years were assessed for body composition parameters (fat, muscle, glycogen, protein masses, bone masses, and body water compartments) and phase angles. PCOS patients demonstrated lower muscle, glycogen and protein masses (U = 60, p = 0.003), along with a lower bone mineral content and mass (U = 78, p = 0.021; U = 74, p = 0.014) than their healthy counterparts, while total and abdominal fat masses were similar between the two groups. PCOS patients also exhibited increased extracellular body water (U = 10, p < 0.001) and decreased intracellular water, compatible with low-grade inflammation and cellular dehydration. Key differences in body composition between women with PCOS and controls demonstrated an osteosarcopenic body composition phenotype in PCOS patients. A confirmation of these findings in larger studies may render osteosarcopenia management a targeted adjunct therapy in women with PCOS.
Collapse
Affiliation(s)
- Charikleia Stefanaki
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, 11527 Athens, Greece.
| | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children's Hospital, 11527 Athens, Greece
| | - Eleni Kandaraki
- Department of Endocrinology, Diabetes and Metabolism, Hygeia Hospital, 15123 Athens, Greece
| | | | | |
Collapse
|