101
|
Ayeleso AO, Joseph JS, Oguntibeju OO, Mukwevho E. Evaluation of free radical scavenging capacity of methoxy containing-hybrids of thiosemicarbazone-triazole and their influence on glucose transport. BMC Pharmacol Toxicol 2018; 19:84. [PMID: 30522526 PMCID: PMC6282370 DOI: 10.1186/s40360-018-0266-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/01/2018] [Indexed: 01/07/2023] Open
Abstract
Background Diabetes mellitus is a metabolic disease in which the body is unable to produce insulin or respond to insulin production, consequently leading to abnormal metabolism of carbohydrates, lipids and proteins causing elevation of glucose in the blood. Oxidative stress, an imbalance between the production of free radicals and body antioxidant system has been implicated in the pathogenesis of diabetes. Free radicals attack important macromolecules leading to cell damage. Antioxidants are intimately involved in the prevention of damage caused by free radicals. Methods The anti-diabetic effects of hybrid compounds (2a-h) of thiosemicarbazone and triazole containing methoxy groups at C (4) positions were tested against genes involved in glucose metabolism (Glut-4, Mef2a and Nrf-1) using quantitative real time PCR (qPCR). Free radical scavenging capacity (FRAP, TEAC, DPPH and ORAC) of the hybrids was also carried out by using established antioxidant capacity assays. Results From the results, hybrid compounds 2b and 2h showed more pronounced effects in up-regulating diabetes associated genes which are important in the up-regulation of glucose uptake. All the hybrid compounds also showed free radical scavenging abilities. Conclusion In conclusion, hybrid compounds (2b and 2h) can be useful as potential drugs for the management of diabetes mellitus.
Collapse
Affiliation(s)
- Ademola O Ayeleso
- Department of Biochemistry, North-West University, Mmabatho, 2735, South Africa.,Department of Biochemistry, Adeleke University, Ede, Osun State, P.M.B. 250, Nigeria
| | - Jitcy S Joseph
- Department of Life & Consumer Sciences, University of South Africa, Florida Campus, Johannesburg, 1709, South Africa
| | - Oluwafemi O Oguntibeju
- Oxidative Stress Research Centre, Department of Biomedical Sciences, Cape Peninsula University of Technology, Bellville, 7535, South Africa
| | - Emmanuel Mukwevho
- Department of Biochemistry, North-West University, Mmabatho, 2735, South Africa.
| |
Collapse
|
102
|
Colaianni G, Lippo L, Sanesi L, Brunetti G, Celi M, Cirulli N, Passeri G, Reseland J, Schipani E, Faienza MF, Tarantino U, Colucci S, Grano M. Deletion of the Transcription Factor PGC-1α in Mice Negatively Regulates Bone Mass. Calcif Tissue Int 2018; 103:638-652. [PMID: 30094757 DOI: 10.1007/s00223-018-0459-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/16/2018] [Indexed: 01/05/2023]
Abstract
Peroxisome proliferator-activated receptor-gamma coactivator (PGC1α) is a transcription coactivator that interacts with a broad range of transcription factors involved in several biological responses. Here, we show that PGC1α plays a role in skeletal homeostasis since aged PGC1α-deficient mice (PGC1α-/-) display impaired bone structure. Micro-CT of the tibial mid-shaft showed a marked decrease of cortical thickness in PGC1α-/- (- 11.9%, p < 0.05) mice compared to wild-type littermate. Trabecular bone was also impaired in knock out mice which displayed lower trabecular thickness (Tb.Th) (- 5.9% vs PGC1α+/+, p < 0.05), whereas trabecular number (Tb.N) was higher than wild-type mice (+ 72% vs PGC1α+/+, p < 0.05), thus resulting in increased (+ 31.7% vs PGC1α+/+, p < 0.05) degree of anisotropy (DA), despite unchanged bone volume fraction (BV/TV). Notably, these impairments of cortical and trabecular bone led to a dramatic ~ 48.4% decrease in bending strength (p < 0.01). These changes in PGC1α-/- mice were paralleled by a significant increase in osteoclast number at the cortical bone surface and in serum level of the bone resorption marker, namely, C-terminal cross-linked telopeptides of type I collagen (CTX-I). We also found that in cortical bone, there was lower expression of mRNA codifying for the key bone-building protein Osteocalcin (Ocn). Interestingly, Collagen I mRNA expression was reduced in mesenchymal stem cells from bone marrow of PGC1α-/-, thus indicating that differentiation of osteoblast lineage is downregulated. Overall, results presented herein suggest that PGC1α may play a key role in bone metabolism.
Collapse
Affiliation(s)
- Graziana Colaianni
- Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Luciana Lippo
- Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
- PhD School in Tissue and Organ Transplantation and Cellular Therapies, Department of Emergency and Organ Transplantation, School of Medicine-University of Bari, Bari, Italy
| | - Lorenzo Sanesi
- Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giacomina Brunetti
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Monica Celi
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Nunzio Cirulli
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Giovanni Passeri
- Department of Clinical and Experimental Medicine, University of Parma, Parma, Italy
| | - Janne Reseland
- Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Ernestina Schipani
- Departments of Medicine and Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Maria Felicia Faienza
- Department of Biomedical Science and Human Oncology, Pediatric Unit, University of Bari, Bari, Italy
| | - Umberto Tarantino
- Department of Orthopedics and Traumatology, Tor Vergata University of Rome, Rome, Italy
| | - Silvia Colucci
- Department of Basic Medical Science, Neuroscience and Sense Organs, University of Bari, Bari, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
103
|
Min W, Fang P, Huang G, Shi M, Zhang Z. The decline of whole-body glucose metabolism in ovariectomized rats. Exp Gerontol 2018; 113:106-112. [PMID: 30292771 DOI: 10.1016/j.exger.2018.09.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/18/2018] [Accepted: 09/30/2018] [Indexed: 02/06/2023]
Abstract
Age is a major risk factor for developing chronic diseases, including type 2 diabetes and osteoporosis. Emerging evidences suggest that the disorder of bone metabolism in osteoporosis is involved in the pathogenesis of glucose intolerance, insulin resistance and type 2 diabetes. However, their etiology and relative regulatory factors still remain elusive to clinicians and researchers. In this study, rats were divided into two groups: normal sham surgery control and ovariectomized (OVX) groups. We evaluated the global bone parameters, glucose metabolism, protein and gene expressions in both skeletal muscle and adipocytes. The present findings showed that the bone mineral density (BMD) and compression load of bone were markedly reduced in OVX rats as revealed by micro-CT, dual energy X-ray absorptiometry and bone biomechanics analysis. Besides, plasma estrogen, total alkaline phosphatase (TALP) and osteocalcin levels were significantly decreased in the OVX rats, but body weight, fat mass and plasma tartrate-resistant acid phosphatase (TRAP) and chemerin levels were significantly increased in the OVX rats. More interestingly, we found that p-AKT, p-P38MAPK, glucose transporter 4 (GLUT4) and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α) contents as well as GLUT4 and PGC-1α mRNA expression were significantly decreased in skeletal muscle and adipocytes of OVX rats. In conclusion, our results indicated that whole-body glucose metabolism and glucose intolerance in OVX rats was degressive, suggesting there was a novel link between osteoporosis and whole body glucose homeostasis, which are controlled by the P38MAPK/PGC-1α/GLUT4 signaling pathway.
Collapse
Affiliation(s)
- Wen Min
- Institute of Bone injury of Traditional Chinese Medicine, Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Penghua Fang
- Institute of Bone injury of Traditional Chinese Medicine, Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Age-related Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Guicheng Huang
- Institute of Bone injury of Traditional Chinese Medicine, Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Mingyi Shi
- Institute of Bone injury of Traditional Chinese Medicine, Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| |
Collapse
|
104
|
Dadvar S, Ferreira DMS, Cervenka I, Ruas JL. The weight of nutrients: kynurenine metabolites in obesity and exercise. J Intern Med 2018; 284:519-533. [PMID: 30141532 DOI: 10.1111/joim.12830] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obesity ultimately results from an imbalance between energy intake and expenditure. However, in addition to their bioenergetic value, nutrients and their metabolites can function as important signalling molecules in energy homeostasis. Indeed, macronutrients and their metabolites can be direct regulators of metabolism through their actions on different organs. In turn, target organs can decide to use, store or transform the incoming nutrients depending on their physiological context and in coordination with other cell types. Tryptophan-kynurenine metabolites are an example of a family of compounds that can serve as systemic integrators of energy metabolism by signalling to different cell types. These include adipocytes, immune cells and muscle fibres, in addition to the well-known effects of kynurenine metabolites on the central nervous system. In the context of energy metabolism, several of the effects elicited by kynurenic acid are mediated by the G-protein-coupled receptor, GPR35. As GPR35 is expressed in tissues such as the adipose tissue, immune cells and the gastrointestinal tract, this receptor could be a potential therapeutic target for the treatment of obesity, diabetes and other metabolic diseases. In addition, metabolic disorders often coincide with states of chronic inflammation, which further highlights GPR35 as an integration node in conditions where inflammation skews metabolism. Defining the molecular interplay between different tissues in the regulation of energy homeostasis can help us understand interindividual variability in the response to nutrient intake and develop safe and efficient therapies to fight obesity and metabolic disease.
Collapse
Affiliation(s)
- S Dadvar
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - D M S Ferreira
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - I Cervenka
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - J L Ruas
- Department of Physiology and Pharmacology, Molecular & Cellular Exercise Physiology, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| |
Collapse
|
105
|
Brown EL, Foletta VC, Wright CR, Sepulveda PV, Konstantopoulos N, Sanigorski A, Della Gatta P, Cameron-Smith D, Kralli A, Russell AP. PGC-1α and PGC-1β Increase Protein Synthesis via ERRα in C2C12 Myotubes. Front Physiol 2018; 9:1336. [PMID: 30356878 PMCID: PMC6190860 DOI: 10.3389/fphys.2018.01336] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/04/2018] [Indexed: 12/25/2022] Open
Abstract
The transcriptional coactivators peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) and PGC-1β are positive regulators of skeletal muscle mass and energy metabolism; however, whether they influence muscle growth and metabolic adaptations via increased protein synthesis is not clear. This study revealed PGC-1α or PGC-1β overexpression in C2C12 myotubes increased protein synthesis and myotube diameter under basal conditions and attenuated the loss in protein synthesis following the treatment with the catabolic agent, dexamethasone. To investigate whether PGC-1α or PGC-1β signal through the Akt/mTOR pathway to increase protein synthesis, treatment with the PI3K and mTOR inhibitors, LY294002 and rapamycin, respectively, was undertaken but found unable to block PGC-1α or PGC-1β’s promotion of protein synthesis. Furthermore, PGC-1α and PGC-1β decreased phosphorylation of Akt and the Akt/mTOR substrate, p70S6K. In contrast to Akt/mTOR inhibition, the suppression of ERRα, a major effector of PGC-1α and PGC-1β activity, attenuated the increase in protein synthesis and myotube diameter in the presence of PGC-1α or PGC-1β overexpression. To characterize further the biological processes occurring, gene set enrichment analysis of genes commonly regulated by both PGC-1α and PGC-1β was performed following a microarray screen. Genes were found enriched in metabolic and mitochondrial oxidative processes, in addition to protein translation and muscle development categories. This suggests concurrent responses involving both increased metabolism and myotube protein synthesis. Finally, based on their known function or unbiased identification through statistical selection, two sets of genes were investigated in a human exercise model of stimulated protein synthesis to characterize further the genes influenced by PGC-1α and PGC-1β during physiological adaptive changes in skeletal muscle.
Collapse
Affiliation(s)
- Erin L Brown
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| | - Victoria C Foletta
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| | - Craig R Wright
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| | - Patricio V Sepulveda
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| | | | | | - Paul Della Gatta
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| | | | - Anastasia Kralli
- Department of Physiology, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, Australia
| |
Collapse
|
106
|
Wada T, Ichihashi Y, Suzuki E, Kosuge Y, Ishige K, Uchiyama T, Makishima M, Nakao R, Oishi K, Shimba S. Deletion of Bmal1 Prevents Diet-Induced Ectopic Fat Accumulation by Controlling Oxidative Capacity in the Skeletal Muscle. Int J Mol Sci 2018; 19:E2813. [PMID: 30231537 PMCID: PMC6164026 DOI: 10.3390/ijms19092813] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 01/05/2023] Open
Abstract
Brain and muscle arnt-like protein 1 (BMAL1), is a transcription factor known to regulate circadian rhythm. BMAL1 was originally characterized by its high expression in the skeletal muscle. Since the skeletal muscle is the dominant organ system in energy metabolism, the possible functions of BMAL1 in the skeletal muscle include the control of metabolism. Here, we established that its involvement in the regulation of oxidative capacity in the skeletal muscle. Muscle-specific Bmal1 KO mice (MKO mice) displayed several physiological hallmarks for the increase of oxidative capacity. This included increased energy expenditure and oxygen consumption, high running endurance and resistance to obesity with improved metabolic profiles. Also, the phosphorylation status of AMP-activated protein kinase and its downstream signaling substrate acetyl-CoA carboxylase in the MKO mice were substantially higher than those in the Bmal1flox/flox mice. In addition, biochemical and histological studies confirmed the substantial activation of oxidative fibers in the skeletal muscle of the MKO mice. The mechanism includes the regulation of Cacna1s expression, followed by the activation of calcium-nuclear factor of activated T cells (NFAT) axis. We thus conclude that BMAL1 is a critical regulator of the muscular fatty acid level under nutrition overloading and that the mechanism involves the control of oxidative capacity.
Collapse
Affiliation(s)
- Taira Wada
- Laboratory of Health Science, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Chiba, Funabshi 274-8555, Japan.
| | - Yuya Ichihashi
- Laboratory of Health Science, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Chiba, Funabshi 274-8555, Japan.
| | - Emi Suzuki
- Laboratory of Health Science, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Chiba, Funabshi 274-8555, Japan.
| | - Yasuhiro Kosuge
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Chiba, Funabshi 274-8555, Japan.
| | - Kumiko Ishige
- Laboratory of Pharmacology, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Chiba, Funabshi 274-8555, Japan.
| | - Taketo Uchiyama
- Laboratory of Organic Chemistry, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Chiba, Funabshi 274-8555, Japan.
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, School of Medicine, Nihon University, 30-1 Oyaguchi-Kamicho, Itabashi-ku, Tokyo 173-8610, Japan.
| | - Reiko Nakao
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8566, Japan.
| | - Katsutaka Oishi
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki 305-8566, Japan.
| | - Shigeki Shimba
- Laboratory of Health Science, School of Pharmacy, Nihon University, 7-7-1 Narashinodai, Chiba, Funabshi 274-8555, Japan.
| |
Collapse
|
107
|
Hernandez-Carretero A, Weber N, LaBarge SA, Peterka V, Doan NYT, Schenk S, Osborn O. Cysteine- and glycine-rich protein 3 regulates glucose homeostasis in skeletal muscle. Am J Physiol Endocrinol Metab 2018; 315:E267-E278. [PMID: 29634311 PMCID: PMC6139493 DOI: 10.1152/ajpendo.00435.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Skeletal muscle is the major site of postprandial peripheral glucose uptake, but in obesity-induced insulin-resistant states insulin-stimulated glucose disposal is markedly impaired. Despite the importance of skeletal muscle in regulating glucose homeostasis, the specific transcriptional changes associated with insulin-sensitive vs. -resistant states in muscle remain to be fully elucidated. Herein, using an RNA-seq approach we identified 20 genes differentially expressed in an insulin-resistant state in skeletal muscle, including cysteine- and glycine-rich protein 3 ( Csrp3), which was highly expressed in insulin-sensitive conditions but significantly reduced in the insulin-resistant state. CSRP3 has diverse functional roles including transcriptional regulation, signal transduction, and cytoskeletal organization, but its role in glucose homeostasis has yet to be explored. Thus, we investigated the role of CSRP3 in the development of obesity-induced insulin resistance in vivo. High-fat diet-fed CSRP3 knockout (KO) mice developed impaired glucose tolerance and insulin resistance as well as increased inflammation in skeletal muscle compared with wild-type (WT) mice. CSRP3-KO mice had significantly impaired insulin signaling, decreased GLUT4 translocation to the plasma membrane, and enhanced levels of phospho-PKCα in muscle, which all contributed to reduced insulin-stimulated glucose disposal in muscle in HFD-fed KO mice compared with WT mice. CSRP3 is a highly inducible protein and its expression is acutely increased after fasting. After 24h fasting, glucose tolerance was significantly improved in WT mice, but this effect was blunted in CSRP3-KO mice. In summary, we identify a novel role for Csrp3 expression in skeletal muscle in the development of obesity-induced insulin resistance.
Collapse
Affiliation(s)
| | - Natalie Weber
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Samuel A LaBarge
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, California
| | - Veronika Peterka
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Nhu Y Thi Doan
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Simon Schenk
- Department of Orthopedic Surgery, University of California, San Diego, La Jolla, California
| | - Olivia Osborn
- Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
108
|
Phillips C, Fahimi A. Immune and Neuroprotective Effects of Physical Activity on the Brain in Depression. Front Neurosci 2018; 12:498. [PMID: 30093853 PMCID: PMC6070639 DOI: 10.3389/fnins.2018.00498] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/03/2018] [Indexed: 12/13/2022] Open
Abstract
Physical activity-a lifestyle factor that is associated with immune function, neuroprotection, and energy metabolism-modulates the cellular and molecular processes in the brain that are vital for emotional and cognitive health, collective mechanisms that can go awry in depression. Physical activity optimizes the stress response, neurotransmitter level and function (e.g., serotonergic, noradrenergic, dopaminergic, and glutamatergic), myokine production (e.g., interleukin-6), transcription factor levels and correlates [e.g., peroxisome proliferator-activated receptor C coactivator-1α [PGC-1α], mitochondrial density, nitric oxide pathway activity, Ca2+ signaling, reactive oxygen specie production, and AMP-activated protein kinase [AMPK] activity], kynurenine metabolites, glucose regulation, astrocytic health, and growth factors (e.g., brain-derived neurotrophic factor). Dysregulation of these interrelated processes can effectuate depression, a chronic mental illness that affects millions of individuals worldwide. Although the biogenic amine model has provided some clinical utility in understanding chronic depression, a need remains to better understand the interrelated mechanisms that contribute to immune dysfunction and the means by which various therapeutics mitigate them. Fortunately, convergent evidence suggests that physical activity improves emotional and cognitive function in persons with depression, particularly in those with comorbid inflammation. Accordingly, the aims of this review are to (1) underscore the link between inflammatory correlates and depression, (2) explicate immuno-neuroendocrine foundations, (3) elucidate evidence of neurotransmitter and cytokine crosstalk in depressive pathobiology, (4) determine the immunomodulatory effects of physical activity in depression, (5) examine protocols used to effectuate the positive effects of physical activity in depression, and (6) highlight implications for clinicians and scientists. It is our contention that a deeper understanding of the mechanisms by which inflammation contributes to the pathobiology of depression will translate to novel and more effective treatments, particularly by identifying relevant patient populations that can benefit from immune-based therapies within the context of personalized medicine.
Collapse
Affiliation(s)
- Cristy Phillips
- Physical Therapy, Arkansas State University, Jonesboro, AR, United States
- Physical Therapy, University of Tennessee Health Science Center, Memphis, TN, United States
| | | |
Collapse
|
109
|
Nakajima T, Koide S, Yasuda T, Hasegawa T, Yamasoba T, Obi S, Toyoda S, Nakamura F, Inoue T, Poole DC, Kano Y. Muscle hypertrophy following blood flow-restricted, low-force isometric electrical stimulation in rat tibialis anterior: role for muscle hypoxia. J Appl Physiol (1985) 2018; 125:134-145. [DOI: 10.1152/japplphysiol.00972.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Low-force exercise training with blood flow restriction (BFR) elicits muscle hypertrophy as seen typically after higher-force exercise. We investigated the effects of microvascular hypoxia [i.e., low microvascular O2 partial pressures (P mvO2)] during contractions on muscle hypertrophic signaling, growth response, and key muscle adaptations for increasing exercise capacity. Wistar rats were fitted with a cuff placed around the upper thigh and inflated to restrict limb blood flow. Low-force isometric contractions (30 Hz) were evoked via electrical stimulation of the tibialis anterior (TA) muscle. The P mvO2 was determined by phosphorescence quenching. Rats underwent acute and chronic stimulation protocols. Whereas P mvO2 decreased transiently with 30 Hz contractions, simultaneous BFR induced severe hypoxia, reducing P mvO2 lower than present for maximal (100 Hz) contractions. Low-force electrical stimulation (EXER) induced muscle hypertrophy (6.2%, P < 0.01), whereas control group conditions or BFR alone did not. EXER+BFR also induced an increase in muscle mass (11.0%, P < 0.01) and, unique among conditions studied, significantly increased fiber cross-sectional area in the superficial TA ( P < 0.05). Phosphorylation of ribosomal protein S6 was enhanced by EXER+BFR, as were peroxisome proliferator-activated receptor gamma coactivator-1α and glucose transporter 4 protein levels. Fibronectin type III domain-containing protein 5, cytochrome c oxidase subunit 4, monocarboxylate transporter 1 (MCT1), and cluster of differentiation 147 increased with EXER alone. EXER+BFR significantly increased MCT1 expression more than EXER alone. These data demonstrate that microvascular hypoxia during contractions is not essential for hypertrophy. However, hypoxia induced via BFR may potentiate the muscle hypertrophic response (as evidenced by the increased superficial fiber cross-sectional area) with increased glucose transporter and mitochondrial biogenesis, which contributes to the pleiotropic effects of exercise training with BFR that culminate in an improved capacity for sustained exercise. NEW & NOTEWORTHY We investigated the effects of low microvascular O2 partial pressures (P mvO2) during contractions on muscle hypertrophic signaling and key elements in the muscle adaptation for increasing exercise capacity. Although demonstrating that muscle hypoxia is not obligatory for the hypertrophic response to low-force, electrically induced muscle contractions, the reduced P mvO2 enhanced ribosomal protein S6 phosphorylation and potentiated the hypertrophic response. Furthermore, contractions with blood flow restriction increased oxidative capacity, glucose transporter, and mitochondrial biogenesis, which are key determinants of the pleiotropic effects of exercise training.
Collapse
Affiliation(s)
- Toshiaki Nakajima
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Tochigi, Japan
| | - Seiichiro Koide
- Bioscience and Technology Program, Department of Engineering Science, University of Electro-Communications, Tokyo, Japan
| | - Tomohiro Yasuda
- School of Nursing, Seirei Christopher University, Shizuoka, Japan
| | - Takaaki Hasegawa
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Tochigi, Japan
| | | | - Syotaro Obi
- Department of Cardiovascular Medicine and Research Support Center, Dokkyo Medical University, Tochigi, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Tochigi, Japan
| | - Fumitaka Nakamura
- Third Department of Internal Medicine, Teikyo University Chiba Medical Center, Chiba, Japan
| | - Teruo Inoue
- Department of Cardiovascular Medicine, Dokkyo Medical University and Heart Center, Dokkyo Medical University Hospital, Tochigi, Japan
| | - David C. Poole
- Department of Anatomy, Physiology and Kinesiology, Kansas State University, Manhattan, Kansas
| | - Yutaka Kano
- Bioscience and Technology Program, Department of Engineering Science, University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
110
|
Lund J, Aas V, Tingstad RH, Van Hees A, Nikolić N. Utilization of lactic acid in human myotubes and interplay with glucose and fatty acid metabolism. Sci Rep 2018; 8:9814. [PMID: 29959350 PMCID: PMC6026123 DOI: 10.1038/s41598-018-28249-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/11/2018] [Indexed: 01/05/2023] Open
Abstract
Once assumed only to be a waste product of anaerobe glycolytic activity, lactate is now recognized as an energy source in skeletal muscles. While lactate metabolism has been extensively studied in vivo, underlying cellular processes are poorly described. This study aimed to examine lactate metabolism in cultured human myotubes and to investigate effects of lactate exposure on metabolism of oleic acid and glucose. Lactic acid, fatty acid and glucose metabolism were studied in myotubes using [14C(U)]lactic acid, [14C]oleic acid and [14C(U)]glucose, respectively. Myotubes expressed both the MCT1, MCT2, MCT3 and MCT4 lactate transporters, and lactic acid was found to be a substrate for both glycogen synthesis and lipid storage. Pyruvate and palmitic acid inhibited lactic acid oxidation, whilst glucose and α-cyano-4-hydroxycinnamic acid inhibited lactic acid uptake. Acute addition of lactic acid inhibited glucose and oleic acid oxidation, whereas oleic acid uptake was increased. Pretreatment with lactic acid for 24 h did not affect glucose or oleic acid metabolism. By replacing glucose with lactic acid during the whole culturing period, glucose uptake and oxidation were increased by 2.8-fold and 3-fold, respectively, and oleic acid oxidation was increased 1.4-fold. Thus, lactic acid has an important role in energy metabolism of human myotubes.
Collapse
Affiliation(s)
- Jenny Lund
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Vigdis Aas
- Department of Life Sciences and Health, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Ragna H Tingstad
- Department of Life Sciences and Health, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Alfons Van Hees
- Department of Life Sciences and Health, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Nataša Nikolić
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway.
| |
Collapse
|
111
|
Hawkins LJ, Al-Attar R, Storey KB. Transcriptional regulation of metabolism in disease: From transcription factors to epigenetics. PeerJ 2018; 6:e5062. [PMID: 29922517 PMCID: PMC6005171 DOI: 10.7717/peerj.5062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/04/2018] [Indexed: 12/13/2022] Open
Abstract
Every cell in an individual has largely the same genomic sequence and yet cells in different tissues can present widely different phenotypes. This variation arises because each cell expresses a specific subset of genomic instructions. Control over which instructions, or genes, are expressed is largely controlled by transcriptional regulatory pathways. Each cell must assimilate a huge amount of environmental input, and thus it is of no surprise that transcription is regulated by many intertwining mechanisms. This large regulatory landscape means there are ample possibilities for problems to arise, which in a medical context means the development of disease states. Metabolism within the cell, and more broadly, affects and is affected by transcriptional regulation. Metabolism can therefore contribute to improper transcriptional programming, or pathogenic metabolism can be the result of transcriptional dysregulation. Here, we discuss the established and emerging mechanisms for controling transcription and how they affect metabolism in the context of pathogenesis. Cis- and trans-regulatory elements, microRNA and epigenetic mechanisms such as DNA and histone methylation, all have input into what genes are transcribed. Each has also been implicated in diseases such as metabolic syndrome, various forms of diabetes, and cancer. In this review, we discuss the current understanding of these areas and highlight some natural models that may inspire future therapeutics.
Collapse
Affiliation(s)
- Liam J Hawkins
- Institute of Biochemistry, Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Rasha Al-Attar
- Institute of Biochemistry, Department of Biology, Carleton University, Ottawa, ON, Canada
| | - Kenneth B Storey
- Institute of Biochemistry, Department of Biology, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
112
|
Resveratrol Ameliorates Cardiac Dysfunction by Inhibiting Apoptosis via the PI3K/Akt/FoxO3a Pathway in a Rat Model of Diabetic Cardiomyopathy. J Cardiovasc Pharmacol 2018; 70:184-193. [PMID: 28678055 DOI: 10.1097/fjc.0000000000000504] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The aim of this study was to explore the effect and mechanism of action of resveratrol (RSV) on cardiac function in diabetic cardiomyopathy (DCM). Hyperglycemia-induced apoptosis contributes to the pathogenic changes in DCM. RSV treatment inhibited high glucose-induced apoptosis of neonatal rat ventricular myocytes. Additionally, high glucose decreased cell viability, prevented serine-threonine kinase (Akt) and FoxO3a phosphorylation, and suppressed cytoplasmic translocation of FoxO3a. However, these effects of apoptosis were reversed by 10 μM of RSV. The PI3K inhibitor LY294002 abolished the RSV protective effect in vitro. RSV (5 or 50 mg·kg·d orally for 8 weeks) prevented the deterioration of cardiac function and structural cardiomyopathy in a streptozotocin-induced rat model of diabetes and reduced apoptosis in diabetic myocardium. Furthermore, it restored streptozotocin-impaired phosphorylation of Akt and FoxO3a (p-Akt and p-FoxO3a) and suppressed nuclear translocation of FoxO3a in vivo. Together, these data indicate that RSV has therapeutic potential against DCM by inhibiting apoptosis via the PI3K/Akt/FoxO3a pathway.
Collapse
|
113
|
Petr M, Stastny P, Zajac A, Tufano JJ, Maciejewska-Skrendo A. The Role of Peroxisome Proliferator-Activated Receptors and Their Transcriptional Coactivators Gene Variations in Human Trainability: A Systematic Review. Int J Mol Sci 2018; 19:E1472. [PMID: 29762540 PMCID: PMC5983571 DOI: 10.3390/ijms19051472] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 05/11/2018] [Accepted: 05/12/2018] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The peroxisome proliferator-activated receptors (PPARA, PPARG, PPARD) and their transcriptional coactivators' (PPARGC1A, PPARGC1B) gene polymorphisms have been associated with muscle morphology, oxygen uptake, power output and endurance performance. The purpose of this review is to determine whether the PPARs and/or their coactivators' polymorphisms can predict the training response to specific training stimuli. METHODS In accordance with the Preferred Reporting Items for Systematic Reviews and Meta Analyses, a literature review has been run for a combination of PPARs and physical activity key words. RESULTS All ten of the included studies were performed using aerobic training in general, sedentary or elderly populations from 21 to 75 years of age. The non-responders for aerobic training (VO₂peak increase, slow muscle fiber increase and low-density lipoprotein decrease) are the carriers of PPARGC1A rs8192678 Ser/Ser. The negative responders for aerobic training (decrease in VO₂peak) are carriers of the PPARD rs2267668 G allele. The negative responders for aerobic training (decreased glucose tolerance and insulin response) are subjects with the PPARG rs1801282 Pro/Pro genotype. The best responders to aerobic training are PPARGC1A rs8192678 Gly/Gly, PPARD rs1053049 TT, PPARD rs2267668 AA and PPARG rs1801282 Ala carriers. CONCLUSIONS The human response for aerobic training is significantly influenced by PPARs' gene polymorphism and their coactivators, where aerobic training can negatively influence glucose metabolism and VO₂peak in some genetically-predisposed individuals.
Collapse
Affiliation(s)
- Miroslav Petr
- Faculty of Physical Education and Sport, Charles University, 162 52 Prague, Czech Republic.
| | - Petr Stastny
- Faculty of Physical Education and Sport, Charles University, 162 52 Prague, Czech Republic.
| | - Adam Zajac
- Department of Theory and Practice of Sport, The Jerzy Kukuczka Academy of Physical Education in Katowice, 40-065 Katowice, Poland.
| | - James J Tufano
- Faculty of Physical Education and Sport, Charles University, 162 52 Prague, Czech Republic.
| | | |
Collapse
|
114
|
Fang P, Yu M, Min W, Han S, Shi M, Zhang Z, Bo P. Beneficial effect of baicalin on insulin sensitivity in adipocytes of diet-induced obese mice. Diabetes Res Clin Pract 2018; 139:262-271. [PMID: 29526684 DOI: 10.1016/j.diabres.2018.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/19/2018] [Accepted: 03/02/2018] [Indexed: 01/31/2023]
Abstract
AIMS Although baicalin has been shown to increase glucose uptake and insulin sensitivity in skeletal muscle of mice, there is no literature available about the effect of baicalin on insulin sensitivity in adipocytes of diet-induced obese mice. METHODS In the present study, diet-induced obese mice were given 50 mg/kg baicalin intraperitoneally (i.p.) once a day for 21 days, and 3T3-L1 cells were treated with 100, 200, 400 μM baicalin for 3 h. Then insulin resistance indexes and insulin signal protein levels were examined to elucidate whether baicalin increased glucose uptake and GLUT4 translocation in adipocytes of diet-induced obese mice. RESULTS The present findings showed that administration of baicalin decreased food intake, body weight, HOMA-IR and p-p38 MAPK and pERK levels, but enhanced pAKT and PGC-1α contents, as well as GLUT4 mRNA, PGC-1α mRNA expression in adipocytes, and reversed high fat diet-induced glucose intolerance, hyperglycemia and insulin resistance in diet-induced obese mice. Moreover, baicalin treatment increased GLUT4 concentration in plasma membranes of adipocytes. CONCLUSIONS These data demonstrated that baicalin accelerated GLUT4 translocation from intracellular membrane compartments to plasma membranes in adipocytes. Baicalin plays a significant role in elevation of glucose uptake and insulin sensitivity to promote glucose clearance.
Collapse
Affiliation(s)
- Penghua Fang
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Mei Yu
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Wen Min
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Shiyu Han
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Mingyi Shi
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Ping Bo
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
115
|
Yoshino J, Almeda-Valdes P, Moseley AC, Mittendorfer B, Klein S. Percutaneous muscle biopsy-induced tissue injury causes local endoplasmic reticulum stress. Physiol Rep 2018; 6:e13679. [PMID: 29687616 PMCID: PMC5913661 DOI: 10.14814/phy2.13679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 01/12/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is likely involved in the pathogenesis of metabolic dysfunction in people with obesity and diabetes. Although tissue biopsy is often used to evaluate the presence and severity of ER stress, it is not known whether acute tissue injury‐induced by percutaneous muscle biopsy causes ER stress and its potential downstream effects on markers of inflammation and metabolic function. In this study, we tested the hypothesis that percutaneous biopsy‐induced tissue injury causes ER stress and alters inflammatory and metabolic pathways in skeletal muscle. Vastus lateralis muscle tissue was obtained by percutaneous biopsy at 0600 h and 12 h later from either the contralateral leg (Group 1, n = 6) or at the same site as the initial biopsy (Group 2, n = 6) in women who were overweight. Muscle gene expression of selected markers of ER stress, inflammation, and regulators of glucose and lipid metabolism were determined. Compared with Group 1, muscle gene expression in the second biopsy sample obtained in Group 2 demonstrated marked increases in markers of ER stress (GRP78, XBP1, ATF6) and inflammation (IL6, TNF), and alterations in metabolic regulators (decreased expression of GLUT4 and PPARGC1A and increased expression of FASN). Our results suggest that acute tissue injury induced by percutaneous muscle biopsy causes an integrated local response that involves an induction of ER stress and alterations in markers of inflammation and regulators of glucose and lipid metabolism.
Collapse
Affiliation(s)
- Jun Yoshino
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Paloma Almeda-Valdes
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Anna C Moseley
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Bettina Mittendorfer
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Samuel Klein
- Center for Human Nutrition, Division of Geriatrics and Nutritional Science, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
116
|
Transcriptional coactivator PGC-1α contains a novel CBP80-binding motif that orchestrates efficient target gene expression. Genes Dev 2018; 32:555-567. [PMID: 29654059 PMCID: PMC5959238 DOI: 10.1101/gad.309773.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/09/2018] [Indexed: 12/13/2022]
Abstract
In this study, Cho et al. investigated how PGC-1α, a transcriptional coactivator, functions in the metabolic adaptation of mammalian cells to diverse physiological stresses. They used in vitro binding assays, X-ray crystallography, and immunoprecipitations of mouse myoblast cell lysates to define a previously unknown cap-binding protein 80 (CBP80)-binding motif (CBM) in the C terminus of PGC-1α, thus providing insight into a novel cap-binding protein surveillance mechanism. Although peroxisome proliferator-activated receptor-γ (PPARγ) coactivator 1α (PGC-1α) is a well-established transcriptional coactivator for the metabolic adaptation of mammalian cells to diverse physiological stresses, the molecular mechanism by which it functions is incompletely understood. Here we used in vitro binding assays, X-ray crystallography, and immunoprecipitations of mouse myoblast cell lysates to define a previously unknown cap-binding protein 80 (CBP80)-binding motif (CBM) in the C terminus of PGC-1α. We show that the CBM, which consists of a nine-amino-acid α helix, is critical for the association of PGC-1α with CBP80 at the 5′ cap of target transcripts. Results from RNA sequencing demonstrate that the PGC-1α CBM promotes RNA synthesis from promyogenic genes. Our findings reveal a new conduit between DNA-associated and RNA-associated proteins that functions in a cap-binding protein surveillance mechanism, without which efficient differentiation of myoblasts to myotubes fails to occur.
Collapse
|
117
|
Gravel SP. Deciphering the Dichotomous Effects of PGC-1α on Tumorigenesis and Metastasis. Front Oncol 2018; 8:75. [PMID: 29629336 PMCID: PMC5876244 DOI: 10.3389/fonc.2018.00075] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022] Open
Abstract
Metabolic reprogramming confers cancer cells the ability to grow and survive under nutrient-depleted or stressful microenvironments. The amplification of oncogenes, the loss of tumor suppressors, as well as context- and lineage-specific determinants can converge and profoundly affect the metabolic status of cancer cells. Cumulating evidences suggest that highly glycolytic cells under the influence of oncogenes such as BRAF, or evolving in hypoxic microenvironments, will promote metastasis through modulation of multiple steps of tumorigenesis such as the epithelial-to-mesenchymal transition (EMT). On the contrary, increased reliance on mitochondrial respiration is associated with hyperplasic rather than metastatic disease. The PGC-1α transcriptional coactivator, a master regulator of mitochondrial biogenesis, has recently been shown to exert antimetastatic effects in cancer, notably through inhibition of EMT. Besides, PGC-1α has the opposite role in specific cancer subtypes, in which it appears to provide growth advantages. Thus, the regulation and role of PGC-1α in cancer is not univocal, and its use as a prognostic marker appears limited given its highly dynamic nature and its multifaceted regulation by transcriptional and posttranslational mechanisms. Herein, we expose key oncogenic and lineage-specific modules that finely regulate PGC-1α to promote or dampen the metastatic process. We propose a unifying model based on the systematic analysis of its controversial implication in cancer from cell proliferation to EMT and metastasis. This short review will provide a good understanding of current challenges associated with the study of PGC-1α.
Collapse
Affiliation(s)
- Simon-Pierre Gravel
- Laboratory of Metabolic Immunopharmacology, Faculty of Pharmacy, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
118
|
Zhu L, Huang Q, Xie Z, Kang M, Ding H, Chen B, Chen Y, Liu C, Wang Y, Tang W. PPARGC1A rs3736265 G>A polymorphism is associated with decreased risk of type 2 diabetes mellitus and fasting plasma glucose level. Oncotarget 2018; 8:37308-37320. [PMID: 28418876 PMCID: PMC5514910 DOI: 10.18632/oncotarget.16307] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/13/2017] [Indexed: 12/21/2022] Open
Abstract
It has been reported that peroxisome proliferator-activated receptor gamma (PPARG) and peroxisome proliferator-activated receptor gamma co-activator 1 (PPARGC1) family (e.g. PPARGC1A and PPARGC1B) are key agents in the development and pathophysiology of type 2 diabetes mellitus (T2DM). In this study, we designed a case-control study and selected PPARG rs1801282 C>G, PPARG rs3856806 C>T, PPARGC1A rs8192678 C>T, PPARGC1A rs2970847 C>T, PPARGC1A rs3736265 G>A, PPARGC1B rs7732671 G>C and PPARGC1B rs17572019 G>A polymorphisms to assess the relationship between these polymorphisms and T2DM using the SNPscan method. A total of 502 T2DM patients and 784 non-diabetic controls were enrolled. We found that PPARGC1A rs3736265 G>A polymorphism was correlated with a borderline decreased susceptibility of T2DM. In a subgroup analysis by age, sex, alcohol use, smoking status and body mass index, a significantly decreased risk of T2DM in <65 years and female groups was found. Haplotype comparison analysis indicated that CTTCGGG and CTCTGGG haplotypes with the order of PPARG rs1801282 C>G, PPARG rs3856806 C>T, PPARGC1A rs8192678 C>T, PPARGC1A rs2970847 C>T, PPARGC1A rs3736265 G>A, PPARGC1B rs7732671 G>C and PPARGC1B rs17572019 G>A polymorphisms in gene position significantly increased the risk of T2DM. However, CCCCACA haplotype conferred a decreased risk to T2DM. We also found that PPARGC1A rs3736265 A allele decreased the level of fasting plasma glucose (FPG), while increased the level of Triglyceride. In conclusion, Our findings suggest that variants of PPARGC1A rs3736265 G>A polymorphism decrease the level of FPG, improving the expectation of study in individual's prevention strategies to T2DM.
Collapse
Affiliation(s)
- Li Zhu
- Department of Nephrology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Qiuyu Huang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Zhiqiang Xie
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Hao Ding
- Department of Respiratory Disease, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Boyang Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Yu Chen
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, Fujian Province, China
| | - Chao Liu
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, China
| | - Yafeng Wang
- Department of Cardiology, The People's Hospital of Xishuangbanna Dai Autonomous Prefecture, Jinghong, Yunnan Province, China
| | - Weifeng Tang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| |
Collapse
|
119
|
Fang P, Yu M, Min W, Wan D, Han S, Shan Y, Wang R, Shi M, Zhang Z, Bo P. Effect of baicalin on GLUT4 expression and glucose uptake in myotubes of rats. Life Sci 2018; 196:156-161. [PMID: 29459024 DOI: 10.1016/j.lfs.2018.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/07/2018] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
Abstract
AIMS Although baicalin could attenuate obesity-induced insulin resistance, the detailed mechanism of baicalin on glucose uptake has not been sufficiently explored as yet. The aim of this study was to survey if baicalin might facilitate glucose uptake and to explore its signal mechanisms in L6 myotubes. MATERIALS AND METHODS L6 myotubes were treated with 100, 200, 400 μM baicalin for 6 h, 12 h and 24 h in this study. Then 2-NBDG and insulin signal protein levels in myotubes of L6 cells were examined. KEY FINDINGS We discovered that administration of baicalin enhanced GLUT4, PGC-1α, pP38MAPK, pAKT and pAS160 contents, as well as GLUT4 mRNA and PGC-1α mRNA levels in L6 myotubes. The beneficial metabolic changes elicited by baicalin were abrogated in myotubes of L6 by P38MAPK or AKT inhibitors. SIGNIFICANCE These results suggest that baicalin promoted glucose uptake in myotubes by differential regulation on P38MAPK and AKT activity. In conclusion, these data provide insight that baicalin is a powerful and promising agent for the treament of hyperglycemia via AKT/AS160/GLUT4 and P38MAPK/PGC1α/GLUT4 pathway.
Collapse
Affiliation(s)
- Penghua Fang
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China; Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Mei Yu
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Wen Min
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Dan Wan
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Shiyu Han
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Yizhi Shan
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Rui Wang
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Mingyi Shi
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu 225300, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Ping Bo
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China.
| |
Collapse
|
120
|
Pettersson-Klein AT, Izadi M, Ferreira DMS, Cervenka I, Correia JC, Martinez-Redondo V, Southern M, Cameron M, Kamenecka T, Agudelo LZ, Porsmyr-Palmertz M, Martens U, Lundgren B, Otrocka M, Jenmalm-Jensen A, Griffin PR, Ruas JL. Small molecule PGC-1α1 protein stabilizers induce adipocyte Ucp1 expression and uncoupled mitochondrial respiration. Mol Metab 2018; 9:28-42. [PMID: 29428596 PMCID: PMC5870114 DOI: 10.1016/j.molmet.2018.01.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 01/12/2018] [Accepted: 01/19/2018] [Indexed: 11/22/2022] Open
Abstract
Objective The peroxisome proliferator-activated receptor-γ coactivator-1α1 (PGC-1α1) regulates genes involved in energy metabolism. Increasing adipose tissue energy expenditure through PGC-1α1 activation is potentially beneficial for systemic metabolism. Pharmacological PGC-1α1 activators could be valuable tools in the fight against obesity and metabolic disease. Finding such compounds has been challenging partly because PGC-1α1 is a transcriptional coactivator with no known ligand-binding properties. While, PGC-1α1 activation is regulated by several mechanisms, protein stabilization is a crucial limiting step due to its short half-life under unstimulated conditions. Methods We designed a cell-based high-throughput screening system to identify PGC-1α1 protein stabilizers. Positive hits were tested for their ability to induce endogenous PGC-1α1 protein accumulation and activate target gene expression in brown adipocytes. Select compounds were analyzed for their effects on global gene expression and cellular respiration in adipocytes. Results Among 7,040 compounds screened, we highlight four small molecules with high activity as measured by: PGC-1α1 protein accumulation, target gene expression, and uncoupled mitochondrial respiration in brown adipocytes. Conclusions We identify compounds that induce PGC-1α1 protein accumulation and show that this increases uncoupled respiration in brown adipocytes. This screening platform establishes the foundation for a new class of therapeutics with potential use in obesity and associated disorders. A high-throughput platform to identify PGC-1α1 activators. PGC-1α1 protein stabilizers work as activators in brown adipocytes. Small molecule PGC-1α1 activators induce Ucp1 expression and cellular respiration.
Collapse
Affiliation(s)
- A T Pettersson-Klein
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - M Izadi
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - D M S Ferreira
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - I Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - J C Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - V Martinez-Redondo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - M Southern
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - M Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - T Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - L Z Agudelo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - M Porsmyr-Palmertz
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - U Martens
- Science for Life Laboratory, RNAi Cell Screening Facility, Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden
| | - B Lundgren
- Science for Life Laboratory, RNAi Cell Screening Facility, Department of Biochemistry and Biophysics, Stockholm University, S-106 91 Stockholm, Sweden
| | - M Otrocka
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - A Jenmalm-Jensen
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - P R Griffin
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, FL, USA
| | - J L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, SE-171 77 Stockholm, Sweden.
| |
Collapse
|
121
|
Amoasii L, Olson EN, Bassel-Duby R. Control of Muscle Metabolism by the Mediator Complex. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a029843. [PMID: 28432117 DOI: 10.1101/cshperspect.a029843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Exercise represents an energetic challenge to whole-body homeostasis. In skeletal muscle, exercise activates a variety of signaling pathways that culminate in the nucleus to regulate genes involved in metabolism and contractility; however, much remains to be learned about the transcriptional effectors of exercise. Mediator is a multiprotein complex that links signal-dependent transcription factors and other transcriptional regulators with the basal transcriptional machinery, thereby serving as a transcriptional "hub." In this article, we discuss recent studies highlighting the role of Mediator subunits in metabolic regulation and glucose metabolism, as well as exercise responsiveness. Elucidation of the roles of Mediator subunits in metabolic control has revealed new mechanisms and molecular targets for the modulation of metabolism and metabolic disorders.
Collapse
Affiliation(s)
- Leonela Amoasii
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 7539-9148
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 7539-9148
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 7539-9148
| |
Collapse
|
122
|
Islam H, Edgett BA, Gurd BJ. Coordination of mitochondrial biogenesis by PGC-1α in human skeletal muscle: A re-evaluation. Metabolism 2018; 79:42-51. [PMID: 29126696 DOI: 10.1016/j.metabol.2017.11.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/13/2017] [Accepted: 11/01/2017] [Indexed: 02/07/2023]
Abstract
The transcriptional co-activator peroxisome proliferator-activated receptor gamma co-activator-1 alpha (PGC-1α) is proposed to coordinate skeletal muscle mitochondrial biogenesis through the integrated induction of nuclear- and mitochondrial-encoded gene transcription. This paradigm is based largely on experiments demonstrating PGC-1α's ability to co-activate various nuclear transcription factors that increase the expression of mitochondrial genes, as well as PGC-1α's direct interaction with mitochondrial transcription factor A within mitochondria to increase the transcription of mitochondrial DNA. While this paradigm is supported by evidence from cellular and transgenic animal models, as well as acute exercise studies involving animals, the up-regulation of nuclear- and mitochondrial-encoded genes in response to exercise does not appear to occur in a coordinated fashion in human skeletal muscle. This review re-evaluates our current understanding of this phenomenon by highlighting evidence from recent studies examining the exercise-induced expression of nuclear- and mitochondrial-encoded genes targeted by PGC-1α. We also highlight several possible theories that may explain the apparent inability of PGC-1α to coordinately up-regulate the expression of genes required for mitochondrial biogenesis in human skeletal muscle, and provide directions for future work exploring mitochondrial biogenic gene expression following exercise.
Collapse
Affiliation(s)
- Hashim Islam
- School of Kinesiology and Health Studies, Queen's University, Kingston K7L 3N6, Ontario, Canada.
| | - Brittany A Edgett
- School of Kinesiology and Health Studies, Queen's University, Kingston K7L 3N6, Ontario, Canada; Human Health and Nutritional Sciences, University of Guelph, Guelph N1G 2W1, Ontario, Canada.
| | - Brendon J Gurd
- School of Kinesiology and Health Studies, Queen's University, Kingston K7L 3N6, Ontario, Canada.
| |
Collapse
|
123
|
Shen L, Meng X, Zhang Z, Wang T. Physical Exercise for Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:529-545. [PMID: 30390268 DOI: 10.1007/978-981-13-1435-3_24] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The most direct characteristic of muscle atrophy is reduction in muscle mass, which is due to increased protein degradation or reduced protein synthesis in skeletal muscle. The loss of muscle mass can directly affect the quality of daily life, prolong the recovery period, and become the main risk factor for chronic diseases. However, there is currently no effective way to prevent and treat this disease, and therefore it is imperative to explore effective therapeutic approaches for muscle atrophy. It is well known that physical exercise is important for maintaining good health and long-term adherence to exercise can reduce the risk of cardiovascular diseases, obesity, and diabetes. It is also well established that exercise training can promote the synthesis of muscle protein and activate signaling pathways that regulate the metabolism and function of muscle fibers. Therefore, exercise can be used as a method to treat muscle atrophy in many of these conditions. Mitochondria play an important role in skeletal muscle homeostasis and bioenergy metabolism. Mitochondria are sensitive to contractile signals, and hence exercise can improve mitochondrial function and promote biosynthesis, which ultimately maintains the healthy state of cells and the whole body. On the other hand, frequent unaccustomed exercise will change the structure and function of skeletal muscle fibers, which is called exercise-induced muscle damage. When the exercise-induced muscle damage happens, it can cause temporary muscle damage and soreness, giving a negative effect on the muscle function.
Collapse
Affiliation(s)
- Liang Shen
- Physical Education College of Shanghai University, Shanghai, China
| | - Xiangmin Meng
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Zhongrong Zhang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China
| | - Tianhui Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, China.
- Shanghai Key Laboratory of Bio-Energy Crops, School of Life Sciences, Shanghai University, Shanghai, China.
| |
Collapse
|
124
|
Consitt LA, Clark BC. The Vicious Cycle of Myostatin Signaling in Sarcopenic Obesity: Myostatin Role in Skeletal Muscle Growth, Insulin Signaling and Implications for Clinical Trials. J Frailty Aging 2018; 7:21-27. [PMID: 29412438 PMCID: PMC6909929 DOI: 10.14283/jfa.2017.33] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The age-related loss of skeletal muscle (sarcopenia) is a major health concern as it is associated with physical disability, metabolic impairments, and increased mortality. The coexistence of sarcopenia with obesity, termed 'sarcopenic obesity', contributes to skeletal muscle insulin resistance and the development of type 2 diabetes, a disease prevalent with advancing age. Despite this knowledge, the mechanisms contributing to sarcopenic obesity remain poorly understood, preventing the development of targeted therapeutics. This article will discuss the clinical and physiological consequences of sarcopenic obesity and propose myostatin as a potential candidate contributing to this condition. A special emphasis will be placed on examining the role of myostatin signaling in impairing both skeletal muscle growth and insulin signaling. In addition, the role of myostatin in regulating muscle-to fat cross talk, further exacerbating metabolic dysfunction in the elderly, will be highlighted. Lastly, we discuss how this knowledge has implications for the design of myostatin-inhibitor clinical trials.
Collapse
Affiliation(s)
- L A Consitt
- Leslie Consitt, PhD, Department of Biomedical Sciences, 228 Irvine Hall, Ohio University, Athens, Ohio, 45701, , Phone: 740-593-2404, Fax: 740-597-2778
| | | |
Collapse
|
125
|
Saleh N, Elayan HE, Zihlif M. THE EFFECT OF SALBUTAMOL ON PGC-1 α AND GLUT4 mRNA EXPRESSION IN THE LIVER AND MUSCLE OF ELDERLY DIABETIC MICE. ACTA ENDOCRINOLOGICA-BUCHAREST 2018; 14:184-191. [PMID: 31149256 DOI: 10.4183/aeb.2018.184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) plays an important role in the regulation of cellular energy metabolism, and it is involved in obesity and type 2 diabetes mellitus (T2DM). Its expression is elevated in the liver of T2DM mouse models. Literature reports show that chronic β2 stimulation improved insulin sensitivity in T2DM. Objectives We aimed to test the hypotheses that chronic β2 stimulation-induced improvement in insulin sensitivity involves changes in the expression of PGC-1α and glucose transporter 4 (GLUT4). Animals and Methods We fed a locally inbred, 8 months old mice, a high fat diet (HFD) to induce diabetes. These mice gained weight and became insulin resistant. The β2 agonist salbutamol had a beneficial effect on both glucose tolerance and insulin sensitivity after 4 weeks. Results Salbutamol beneficial effect persisted after 4 weeks of its discontinuation. HFD caused an up regulation of the hepatic PGC-1 α expression by 5.23 folds (P< 0.041) and salbutamol reversed this effect and caused a down regulation by 30.3 folds (P< 0.0001). PGC-1 α and GLUT4 expression in the muscle was not affected by salbutamol (P> 0.05). Conclusion Down regulation of the liver's PGC-1 α contributes to the beneficial effect of the chronic β2 stimulation on glucose tolerance and insulin sensitivity in T2DM mice.
Collapse
Affiliation(s)
- N Saleh
- University of Jordan, Faculty of Medicine, Department of Pharmacology, Amman, Jordan
| | - H E Elayan
- University of Jordan, Faculty of Medicine, Department of Pharmacology, Amman, Jordan
| | - M Zihlif
- University of Jordan, Faculty of Medicine, Department of Pharmacology, Amman, Jordan
| |
Collapse
|
126
|
Fang P, Zhang L, Yu M, Sheng Z, Shi M, Zhu Y, Zhang Z, Bo P. Activiated galanin receptor 2 attenuates insulin resistance in skeletal muscle of obese mice. Peptides 2018; 99:92-98. [PMID: 29183756 DOI: 10.1016/j.peptides.2017.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 01/28/2023]
Abstract
The results of our and other's studies showed that activation of galanin receptor 1 could mitigate insulin resistance via promoting glucose transporter 4 (GLUT4) expression and translocation in the skeletal muscle of rats. But no literature are available regarding the effect of galanin receptor 2 (GALR2) on insulin resistance in skeletal muscle of type 2 diabetes. Herein, in this study we intended to survey the effect of GALR2 and its signal mechanisms in the mice with high fat diet-induced obese. The mice were intraperitoneally injected with vehicle, GALR2 agonist M1145 and antagonist M871 respectively once a day for continuous 21 days. The skeletal muscles were processed for determination of glucose uptake, and GLUT4 mRNA and protein expression levels. The PGC-1α, AKT, p38MAPK, AS160, pAKT, pP38MAPK and pAS160 expression levels were quantitatively assessed too. We found that pharmacological activation of GALR2 enhanced energy expenditure, and increased GLUT4 expression and translocation in skeletal muscle of mice during high-fat diet regimens. Activation of GALR2 alleviated insulin resistance through P38MAPK/PGC-1α/GLUT4 and AKT/AS160/GLUT4 pathway in the skeletal muscle of mice. Overall, these results identify that GALR2 is a regulator of insulin resistance and activation of GALR2 represents a promising strategy against obesity-induced insulin resistance.
Collapse
Affiliation(s)
- Penghua Fang
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu, 225300, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Mei Yu
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu, 225300, China
| | - Zhongqi Sheng
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Mingyi Shi
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Yan Zhu
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China.
| | - Ping Bo
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
127
|
de Souza JFT, Dáttilo M, de Mello MT, Tufik S, Antunes HKM. High-Intensity Interval Training Attenuates Insulin Resistance Induced by Sleep Deprivation in Healthy Males. Front Physiol 2017; 8:992. [PMID: 29270126 PMCID: PMC5725446 DOI: 10.3389/fphys.2017.00992] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/20/2017] [Indexed: 01/08/2023] Open
Abstract
Introduction: Sleep deprivation can impair several physiological systems and recently, new evidence has pointed to the relationship between a lack of sleep and carbohydrate metabolism, consequently resulting in insulin resistance. To minimize this effect, High-Intensity Interval Training (HIIT) is emerging as a potential strategy. Objective: The aim of this study was to investigate the effects of HIIT on insulin resistance induced by sleep deprivation. Method: Eleven healthy male volunteers were recruited, aged 18–35 years, who declared taking 7–8 h sleep per night. All volunteers were submitted to four different conditions: a single night of regular sleep (RS condition), 24 h of total sleep deprivation (SD condition), HIIT training followed by regular sleep (HIIT+RS condition), and HIIT training followed by 24 h of total sleep deprivation (HIIT+SD condition). They performed six training sessions over 2 weeks and each session consisted of 8–12 × 60 s intervals at 100% of peak power output. In each experimental condition, tests for glucose, insulin, cortisol, free fatty acids, and insulin sensitivity, measured by oral glucose tolerance test (OGTT), were performed. Results: Sleep deprivation increased glycaemia and insulin levels, as well as the area under the curve. Furthermore, an increase in free fatty acids concentrations and basal metabolism was observed. There were no differences in the concentrations of cortisol. However, HIIT before 24 h of sleep deprivation attenuated the increase of glucose, insulin, and free fatty acids. Conclusion: Twenty-four hours of sleep deprivation resulted in acute insulin resistance. However, HIIT is an effective strategy to minimize the deleterious effects promoted by this condition.
Collapse
Affiliation(s)
- Jorge F T de Souza
- Departamento de Biociências, Universidade Federal de São Paulo, São Paulo, Brazil.,Centro de Estudos em Psicobiologia e Exercício, São Paulo, Brazil
| | - Murilo Dáttilo
- Centro de Estudos em Psicobiologia e Exercício, São Paulo, Brazil.,Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marco T de Mello
- Departamento de Esportes, Faculdade de Educação Física, Fisioterapia e Terapia Ocupacional, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sergio Tufik
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Hanna K M Antunes
- Departamento de Biociências, Universidade Federal de São Paulo, São Paulo, Brazil.,Centro de Estudos em Psicobiologia e Exercício, São Paulo, Brazil
| |
Collapse
|
128
|
Schnuck JK, Gould LM, Parry HA, Johnson MA, Gannon NP, Sunderland KL, Vaughan RA. Metabolic effects of physiological levels of caffeine in myotubes. J Physiol Biochem 2017; 74:35-45. [PMID: 29198059 DOI: 10.1007/s13105-017-0601-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022]
Abstract
Caffeine has been shown to stimulate multiple major regulators of cell energetics including AMP-activated protein kinase (AMPK) and Ca2+/calmodulin-dependent protein kinase II (CaMKII). Additionally, caffeine induces peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and mitochondrial biogenesis. While caffeine enhances oxidative metabolism, experimental concentrations often exceed physiologically attainable concentrations through diet. This work measured the effects of low-level caffeine on cellular metabolism and gene expression in myotubes, as well as the dependence of caffeine's effects on the nuclear receptor peroxisome proliferator-activated receptor beta/delta (PPARβ/δ). C2C12 myotubes were treated with various doses of caffeine for up to 24 h. Gene and protein expression were measured via qRT-PCR and Western blot, respectively. Cellular metabolism was determined via oxygen consumption and extracellular acidification rate. Caffeine significantly induced regulators of mitochondrial biogenesis and oxidative metabolism. Mitochondrial staining was suppressed in PPARβ/δ-inhibited cells which was rescued by concurrent caffeine treatment. Caffeine-treated cells also displayed elevated peak oxidative metabolism which was partially abolished following PPARβ/δ inhibition. Similar to past observations, glucose uptake and GLUT4 content were elevated in caffeine-treated cells, however, glycolytic metabolism was unaltered following caffeine treatment. Physiological levels of caffeine appear to enhance cell metabolism through mechanisms partially dependent on PPARβ/δ.
Collapse
Affiliation(s)
- Jamie K Schnuck
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
- School of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Lacey M Gould
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
| | - Hailey A Parry
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
- School of Kinesiology, Auburn University, Auburn, AL, 36849, USA
| | - Michele A Johnson
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
| | - Nicholas P Gannon
- School of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Kyle L Sunderland
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA
| | - Roger A Vaughan
- Department of Exercise Science, High Point University, One University Parkway, High Point, NC, 27268, USA.
| |
Collapse
|
129
|
Lee J, Salazar Hernández MA, Auen T, Mucka P, Lee J, Ozcan U. PGC-1α functions as a co-suppressor of XBP1s to regulate glucose metabolism. Mol Metab 2017; 7:119-131. [PMID: 29129613 PMCID: PMC5784318 DOI: 10.1016/j.molmet.2017.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/15/2017] [Accepted: 10/24/2017] [Indexed: 12/12/2022] Open
Abstract
Objective Peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) promotes hepatic gluconeogenesis by activating HNF4α and FoxO1. PGC-1α expression in the liver is highly elevated in obese and diabetic conditions, leading to increased hepatic glucose production. We previously showed that the spliced form of X-box binding protein 1 (XBP1s) suppresses FoxO1 activity and hepatic gluconeogenesis. The shared role of PGC-1α and XBP1s in regulating FoxO1 activity and gluconeogenesis led us to investigate the probable interaction between PGC-1α and XBP1s and its role in glucose metabolism. Methods We investigated the biochemical interaction between PGC-1α and XBP1s and examined the role of their interaction in glucose homeostasis using animal models. Results We show that PGC-1α interacts with XBP1s, which plays an anti-gluconeogenic role in the liver by suppressing FoxO1 activity. The physical interaction between PGC-1α and XBP1s leads to suppression of XBP1s activity rather than its activation. Upregulating PGC-1α expression in the liver of lean mice lessens XBP1s protein levels, and reducing PGC-1α levels in obese and diabetic mouse liver restores XBP1s protein induction. Conclusions Our findings reveal a novel function of PGC-1α as a suppressor of XBP1s function, suggesting that hepatic PGC-1α promotes gluconeogenesis through multiple pathways as a co-activator for HNF4α and FoxO1 and also as a suppressor for anti-gluconeogenic transcription factor XBP1s. XBP1s suppresses FoxO1 activity and hepatic gluconeogenesis. PGC-1α physically interacts with XBP1s and functions as a co-suppressor of XBP1s in the liver. The suppression of XBP1s activity by PGC-1α leads to impaired glucose homeostasis in obese mice. Hepatic PGC-1α promotes gluconeogenesis as a co-activator and also as a co-suppressor.
Collapse
Affiliation(s)
- Jaemin Lee
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Current address: Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| | | | - Thomas Auen
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Mucka
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Justin Lee
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Umut Ozcan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
130
|
Tobina T, Mori Y, Doi Y, Nakayama F, Kiyonaga A, Tanaka H. Peroxisome proliferator-activated receptor gamma co-activator 1 gene Gly482Ser polymorphism is associated with the response of low-density lipoprotein cholesterol concentrations to exercise training in elderly Japanese. J Physiol Sci 2017; 67:595-602. [PMID: 27699582 PMCID: PMC10717479 DOI: 10.1007/s12576-016-0491-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/20/2016] [Indexed: 01/20/2023]
Abstract
Muscle peroxisome proliferator-activated receptor gamma co-activator 1 (PGC-1)α gene expression is influenced by the Gly482Ser gene polymorphism, which is a candidate genetic risk factor for diabetes mellitus and obesity. This study investigated the effects of PGC-1 gene Gly482Ser polymorphisms on alterations in glucose and lipid metabolism induced by exercise training. A 12-week intervention study was performed for 119 participants who were more than 65 years of age and completed exercise training at lactate threshold intensity. Total cholesterol and low-density lipoprotein cholesterol were significantly reduced in Gly/Gly but not in Gly/Ser and Ser/Ser participants after exercise. The Gly/Gly genotype of the PGC-1 gene Gly482Ser polymorphism influences the effects of moderate-intensity exercise training on low-density lipoprotein cholesterol and total cholesterol concentrations in older people.
Collapse
Affiliation(s)
- Takuro Tobina
- Faculty of Nursing and Nutrition, University of Nagasaki, Manabino1-1-1, Nagayo-cho, Nishisonogi-gun, Nagasaki, 851-2195, Japan.
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan.
| | - Yukari Mori
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| | - Yukiko Doi
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Commerce, Department of Tourism Industry, Kyushu Sangyo University, Fukuoka, Japan
| | - Fuki Nakayama
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| | - Akira Kiyonaga
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| | - Hiroaki Tanaka
- The Fukuoka University Institute for Physical Activity, Fukuoka University, Fukuoka, Japan
- Faculty of Sports Science, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
131
|
Miyashita K, Hosokawa M. Fucoxanthin in the management of obesity and its related disorders. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
132
|
Ribosomal transcription is regulated by PGC-1alpha and disturbed in Huntington's disease. Sci Rep 2017; 7:8513. [PMID: 28819135 PMCID: PMC5561056 DOI: 10.1038/s41598-017-09148-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 07/24/2017] [Indexed: 12/12/2022] Open
Abstract
PGC-1α is a versatile inducer of mitochondrial biogenesis and responsive to the changing energy demands of the cell. As mitochondrial ATP production requires proteins that derive from translation products of cytosolic ribosomes, we asked whether PGC-1α directly takes part in ribosomal biogenesis. Here, we show that a fraction of cellular PGC-1α localizes to the nucleolus, the site of ribosomal transcription by RNA polymerase I. Upon activation PGC-1α associates with the ribosomal DNA and boosts recruitment of RNA polymerase I and UBF to the rDNA promoter. This induces RNA polymerase I transcription under different stress conditions in cell culture and mouse models as well as in healthy humans and is impaired already in early stages of human Huntington’s disease. This novel molecular link between ribosomal and mitochondrial biogenesis helps to explain sarcopenia and cachexia in diseases of neurodegenerative origin.
Collapse
|
133
|
Woelfel JR, Kimball AL, Yen CL, Shields RK. Low-Force Muscle Activity Regulates Energy Expenditure after Spinal Cord Injury. Med Sci Sports Exerc 2017; 49:870-878. [PMID: 28009786 DOI: 10.1249/mss.0000000000001187] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Reduced physical activity is a primary risk factor for increased morbidity and mortality. People with spinal cord injury (SCI) have reduced activity for a lifetime, as they cannot volitionally activate affected skeletal muscles. We explored whether low-force and low-frequency stimulation is a viable strategy to enhance systemic energy expenditure in people with SCI. PURPOSE This study aimed to determine the effects of low stimulation frequency (1 and 3 Hz) and stimulation intensity (50 and 100 mA) on energy expenditure in people with SCI. We also examined the relationship between body mass index and visceral adipose tissue on energy expenditure during low-frequency stimulation. METHODS Ten individuals with complete SCI underwent oxygen consumption monitoring during electrical activation of the quadriceps and hamstrings at 1 and 3 Hz and at 50 and 100 mA. We calculated the difference in energy expenditure between stimulation and rest and estimated the number of days that would be necessary to burn 1 lb of body fat (3500 kcal) for each stimulation protocol (1 vs 3 Hz). RESULTS Both training frequencies induced a significant increase in oxygen consumption above a resting baseline level (P < 0.05). Energy expenditure positively correlated with stimulus intensity (muscle recruitment) and negatively correlated with adiposity (reflecting the insulating properties of adipose tissue). We estimated that 1 lb of body fat could be burned more quickly with 1 Hz training (58 d) as compared with 3 Hz training (87 d) if an identical number of pulses were delivered. CONCLUSION Low-frequency stimulation increased energy expenditure per pulse and may be a feasible option to subsidize physical activity to improve metabolic status after SCI.
Collapse
Affiliation(s)
- Jessica R Woelfel
- 1Carver College of Medicine, University of Iowa, Iowa City, IA; and 2Department of Physical Therapy and Rehabilitation Science, Carver College of Medicine, University of Iowa, Iowa City, IA
| | | | | | | |
Collapse
|
134
|
Resveratrol Improves Glycemic Control in Type 2 Diabetic Obese Mice by Regulating Glucose Transporter Expression in Skeletal Muscle and Liver. Molecules 2017; 22:molecules22071180. [PMID: 28708105 PMCID: PMC6152102 DOI: 10.3390/molecules22071180] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/05/2017] [Accepted: 07/06/2017] [Indexed: 01/10/2023] Open
Abstract
Insulin resistance participates in the glycaemic control disruption in type 2 diabetes mellitus (T2DM), by reducing muscle glucose influx and increasing liver glucose efflux. GLUT4 (Slc2a4 gene) and GLUT2 (Slc2a2 gene) proteins play a fundamental role in the muscle and liver glucose fluxes, respectively. Resveratrol is a polyphenol suggested to have an insulin sensitizer effect; however, this effect, and related mechanisms, have not been clearly demonstrated in T2DM. We hypothesized that resveratrol can improve glycaemic control by restoring GLUT4 and GLUT2 expression in muscle and liver. Mice were rendered obese T2DM in adult life by neonatal injection of monosodium glutamate. Then, T2DM mice were treated with resveratrol for 60 days or not. Glycaemic homeostasis, GLUT4, GLUT2, and SIRT1 (sirtuin 1) proteins (Western blotting); Slc2a4, Slc2a2, and Pck1 (key gluconeogenic enzyme codifier) mRNAs (RT-qPCR); and hepatic glucose efflux were analysed. T2DM mice revealed: high plasma concentration of glucose, fructosamine, and insulin; insulin resistance (insulin tolerance test); decreased Slc2a4/GLUT4 content in gastrocnemius and increased Slc2a2/GLUT2 content in liver; and increased Pck1 mRNA and gluconeogenic activity (pyruvate tolerance test) in liver. All alterations were restored by resveratrol treatment. Additionally, in both muscle and liver, resveratrol increased SIRT1 nuclear content, which must participate in gene expression regulations. In sum, the results indisputably reveals that resveratrol improves glycaemic control in T2DM, and that involves an increase in muscle Slc2a4/GLUT4 and a decrease in liver Slc2a2/GLUT2 expression. This study contributes to our understanding how resveratrol might be prescribed for T2DM according to the principles of evidence-based medicine.
Collapse
|
135
|
Multi-regulatory network of ROS: the interconnection of ROS, PGC-1 alpha, and AMPK-SIRT1 during exercise. J Physiol Biochem 2017; 73:487-494. [PMID: 28707280 DOI: 10.1007/s13105-017-0576-y] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/29/2017] [Indexed: 01/20/2023]
|
136
|
Fang P, Yu M, Zhang L, Wan D, Shi M, Zhu Y, Bo P, Zhang Z. Baicalin against obesity and insulin resistance through activation of AKT/AS160/GLUT4 pathway. Mol Cell Endocrinol 2017; 448:77-86. [PMID: 28359800 DOI: 10.1016/j.mce.2017.03.027] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/04/2017] [Accepted: 03/26/2017] [Indexed: 01/24/2023]
Abstract
Obesity may cause several metabolic complications, including insulin resistance and type 2 diabetes mellitus. Despite great advances in medicine, people still keep exploring novel and effective drugs for treatment of obesity and insulin resistance. The aim of this study was to survey if baicalin might ameliorate obesity-induced insulin resistance and to explore its signal mechanisms in skeletal muscles of mice. Diet-induced obese (DIO) mice were given 50 mg/kg baicalin intraperitoneally (i.p.) once a day for 21 days, and C2C12 myotubes were treated with 100, 200, 400 μM baicalin for 12 h in this study. Then insulin resistance indexes and insulin signal protein levels in skeletal muscles were examined. We discovered that administration of baicalin decreased food intake, body weight, HOMA-IR and NT-PGC-1α levels, but enhanced GLUT4, PGC-1α, pP38MAPK, pAKT and pAS160 contents, as well as GLUT4 mRNA, PGC-1α mRNA, PPARγ mRNA, GLUT1 mRNA expression in skeletal muscles of obese mice and myotubes of C2C12 cells, and reversed high fat diet-induced glucose and insulin intolerance, hyperglycemia and insulin resistance in the mice. These results suggest that baicalin is a powerful and promising agent for treatment of obesity and insulin resistance via Akt/AS160/GLUT4 and P38MAPK/PGC1α/GLUT4 pathway.
Collapse
Affiliation(s)
- Penghua Fang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu, 225300, China
| | - Mei Yu
- Department of Physiology, Nanjing University of Chinese Medicine Hanlin College, Taizhou, Jiangsu, 225300, China
| | - Lei Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Dan Wan
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Mingyi Shi
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China
| | - Yan Zhu
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
| | - Ping Bo
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou 225001, China; Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China.
| | - Zhenwen Zhang
- Department of Endocrinology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China.
| |
Collapse
|
137
|
Ibrahim A, Neinast M, Arany ZP. Myobolites: muscle-derived metabolites with paracrine and systemic effects. Curr Opin Pharmacol 2017; 34:15-20. [PMID: 28441626 PMCID: PMC5651206 DOI: 10.1016/j.coph.2017.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 02/24/2017] [Accepted: 03/06/2017] [Indexed: 02/08/2023]
Abstract
Intracellular metabolism in skeletal muscle has been studied for more than a century and is the stuff of textbooks. In contrast, the extracellular secretion of metabolites by muscle cells, and their effects on non-muscle cells near or far, has been investigated much less extensively. Here, we describe a number of cases in which striated muscle secretes a metabolite that elicits complex responses in other cells or tissues, with involvements in normal physiology as well as obesity, type II diabetes, and cardiac remodeling. We focus on two recently identified secreted catabolic products of branched chain amino acid breakdown, β-aminoisobutyric acid and 3-hydroxyisobutyrate, and discuss common themes of inter-cellular signaling pathways driven by secreted metabolites.
Collapse
Affiliation(s)
- Ayon Ibrahim
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19106, USA
| | - Michael Neinast
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19106, USA
| | - Zoltan P Arany
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19106, USA.
| |
Collapse
|
138
|
Niu B, Ke CQ, Li BH, Li Y, Yi Y, Luo Y, Shuai L, Yao S, Lin LG, Li J, Ye Y. Cucurbitane Glucosides from the Crude Extract of Siraitia grosvenorii with Moderate Effects on PGC-1α Promoter Activity. JOURNAL OF NATURAL PRODUCTS 2017; 80:1428-1435. [PMID: 28448136 DOI: 10.1021/acs.jnatprod.6b01086] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Seven new cucurbitane glucosides, 11-oxomogrosides III E and IV (1 and 2), 11-oxoisomogroside V (3), 7-oxomogrosides III E and IV (4 and 5), and mogrosides VI A and VI B (6 and 7), were separated from the crude extract of Siraitia grosvenorii. The new structures were defined by analysis of their 1H and 13C NMR, 2D NMR, and HRESIMS data. Especially, the band-selective constant time HSQC and band-selective constant time HMBC techniques were recuited to elucidate the structures of the complex glucoside moieties. Using the PGC-1α promoter driven luciferase reporter assay, the isolated compounds were examined for PGC-1α promoter activity.
Collapse
Affiliation(s)
- Biao Niu
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
- School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, People's Republic of China
- University of Chinese Academy of Sciences , No. 19A Yuquan Road, Beijing 100049, People's Republic of China
| | - Chang-Qiang Ke
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
| | - Bo-Han Li
- University of Chinese Academy of Sciences , No. 19A Yuquan Road, Beijing 100049, People's Republic of China
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
| | - Yuanyuan Li
- Guilin Layn Natural Ingredients Corp , Guilin 541199, People's Republic of China
| | - Yongji Yi
- Guilin Layn Natural Ingredients Corp , Guilin 541199, People's Republic of China
| | - Yongwei Luo
- Guilin Layn Natural Ingredients Corp , Guilin 541199, People's Republic of China
| | - Lin Shuai
- University of Chinese Academy of Sciences , No. 19A Yuquan Road, Beijing 100049, People's Republic of China
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
| | - Sheng Yao
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
| | - Li-Gen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Avenida da Universidade, Taipa, Macao 999078, People's Republic of China
| | - Jia Li
- University of Chinese Academy of Sciences , No. 19A Yuquan Road, Beijing 100049, People's Republic of China
- Chinese National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 189 Guoshoujing Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
| | - Yang Ye
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , 555 Zu-Chong-Zhi Road, Zhangjiang Hi-Tech Park, Shanghai 201203, People's Republic of China
- School of Life Science and Technology, ShanghaiTech University , Shanghai 201210, People's Republic of China
- University of Chinese Academy of Sciences , No. 19A Yuquan Road, Beijing 100049, People's Republic of China
| |
Collapse
|
139
|
Jaimes-Hoy L, Gurrola GB, Cisneros M, Joseph-Bravo P, Possani LD, Charli JL. The Kv1.3 channel blocker Vm24 enhances muscle glucose transporter 4 mobilization but does not reduce body-weight gain in diet-induced obese male rats. Life Sci 2017; 181:23-30. [PMID: 28549558 DOI: 10.1016/j.lfs.2017.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/14/2017] [Accepted: 05/22/2017] [Indexed: 12/28/2022]
Abstract
AIMS Voltage-gated potassium channels 1.3 (Kv1.3) can be targeted to reduce diet-induced obesity and insulin resistance in mice. Since species-specific differences in Kv1.3 expression and pharmacology have been observed, we tested the effect of Vm24, a high-affinity specific blocker of Kv1.3 channels from Vaejovis mexicanus smithi, on body weight (BW), glucose tolerance and insulin resistance in diet-induced obese rats. MATERIALS AND METHODS Young adult male Wistar rats were switched to a high-fat/high-fructose (HFF) diet. Eighteen days later animals were divided in two groups: vehicle and Vm24 group. Subcutaneous injections were applied every other day until sacrifice 2months later. An additional cohort was maintained on standard chow. KEY FINDINGS The HFF diet promoted obesity. Treatment with Vm24 did not alter various metabolic parameters such as food intake, BW gain, visceral white adipose tissue mass, adipocyte diameter, serum glucose, leptin and thyroid hormone concentrations, brown adipose tissue mass or uncoupling protein-1 expression, and insulin tolerance. Vm24 did reduce basal and glucose-stimulated serum insulin concentrations, serum C-peptide concentration, increased QUICKI, and tended to lower HOMA-IR. Vm24 treatment did not change the activation of insulin receptor substrate-1, but enhanced protein-kinase B activation and membrane glucose-transporter 4 (GLUT4) protein levels in skeletal muscle. SIGNIFICANCE In conclusion, in male rats, long-term blockade of Kv1.3 channels with Vm24 does not reduce weight gain and visceral adiposity induced by HFF diet; instead, it reduces serum insulin concentration, and enhances GLUT4 mobilization in skeletal muscle.
Collapse
Affiliation(s)
- Lorraine Jaimes-Hoy
- Departamento de Fisiología Molecular y Biología del Desarrollo, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Georgina B Gurrola
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Miguel Cisneros
- Departamento de Fisiología Molecular y Biología del Desarrollo, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Patricia Joseph-Bravo
- Departamento de Fisiología Molecular y Biología del Desarrollo, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Lourival D Possani
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Jean-Louis Charli
- Departamento de Fisiología Molecular y Biología del Desarrollo, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
140
|
Jiang S, Teague AM, Tryggestad JB, Chernausek SD. Role of microRNA-130b in placental PGC-1α/TFAM mitochondrial biogenesis pathway. Biochem Biophys Res Commun 2017; 487:607-612. [PMID: 28433632 DOI: 10.1016/j.bbrc.2017.04.099] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 04/18/2017] [Indexed: 11/30/2022]
Abstract
Diabetes during pregnancy is associated with abnormal placenta mitochondrial function and increased oxidative stress, which affect fetal development and offspring long-term health. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis and energy metabolism. The molecular mechanisms underlying the regulation of PGC-1α in placenta in the context of diabetes remain unclear. The present study examined the role of microRNA 130b (miR-130b-3p) in regulating PGC-1α expression and oxidative stress in a placental trophoblastic cell line (BeWo). Prolonged exposure of BeWo cells to high glucose mimicking hyperglycemia resulted in decreased protein abundance of PGC-1α and its downstream factor, mitochondrial transcription factor A (TFAM). High glucose treatment increased the expression of miR-130b-3p in BeWo cells, as well as exosomal secretion of miR-130b-3p. Transfection of BeWo cells with miR-130b-3p mimic reduced the abundance of PGC-1α, whereas inhibition of miR-130b-3p increased PGC-1α expression in response to high glucose, suggesting a role for miR-130b-3p in mediating high glucose-induced down regulation of PGC-1α expression. In addition, miR-130b-3p anti-sense inhibitor increased TFAM expression and reduced 4-hydroxynonenal (4-HNE)-induced production of reactive oxygen species (ROS). Taken together, these findings reveal that miR-130b-3p down-regulates PGC-1α expression in placental trophoblasts, and inhibition of miR-130b-3p appears to improve mitochondrial biogenesis signaling and protect placental trophoblast cells from oxidative stress.
Collapse
Affiliation(s)
- Shaoning Jiang
- Department of Pediatrics, Section of Diabetes and Endocrinology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - April M Teague
- Department of Pediatrics, Section of Diabetes and Endocrinology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jeanie B Tryggestad
- Department of Pediatrics, Section of Diabetes and Endocrinology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Steven D Chernausek
- Department of Pediatrics, Section of Diabetes and Endocrinology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
141
|
Barbosa de Queiroz K, Honorato-Sampaio K, Rossoni Júnior JV, Andrade Leal D, Pinto ABG, Kappes-Becker L, Evangelista EA, Guerra-Sá R. Physical activity prevents alterations in mitochondrial ultrastructure and glucometabolic parameters in a high-sugar diet model. PLoS One 2017; 12:e0172103. [PMID: 28199417 PMCID: PMC5310863 DOI: 10.1371/journal.pone.0172103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/31/2017] [Indexed: 11/19/2022] Open
Abstract
Endurance exercise is a remarkable intervention for the treatment of many diseases. Mitochondrial changes on skeletal muscle are likely important for many of the benefits provided by exercise. In this study, we aimed to evaluate the effects that a regular physical activity (swimming without workload) has on mitochondrial morphological alterations and glucometabolic parameters induced by a high-sugar diet (HSD). Weaned male Wistar rats fed with a standard diet or a HSD (68% carbohydrate) were subjected to 60 minutes of regular physical activity by swimming (without workload) for four- (20 sessions) or eight-week (40 sessions) periods. After training, animals were euthanized and the sera, adipose tissues, and skeletal muscles were collected for further analysis. The HSD increased body weight after an 8-week period; it also increased the fat pads and the adipose index, resulting in glucose intolerance and insulin resistance (IR). Transmission electron microscopy showed an increase in alterations of mitochondrial ultrastructure in the gastrocnemius muscle, as well as a decrease in superoxide dismutase (SOD) activity, and an increase in protein carbonylation. Regular physical activity partially reverted these alterations in rats fed a HSD, preventing mitochondrial morphological alterations and IR. Moreover, we observed a decrease in Pgc1α expression (qPCR analysis) in STD-EXE group and a less pronounced reduction in HSD-EXE group after an 8-week period. Thus, regular physical activity (swimming without workload) in rats fed a HSD can prevent mitochondrial dysfunction and IR, highlighting the crucial role for physical activity on metabolic homeostasis.
Collapse
Affiliation(s)
- Karina Barbosa de Queiroz
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
- * E-mail:
| | - Kinulpe Honorato-Sampaio
- Faculdade de Medicina, Campus JK, Universidade Federal dos Vales Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brasil
| | - Joamyr Victor Rossoni Júnior
- Laboratório de Bioquímica Metabólica, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Diego Andrade Leal
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | | | - Lenice Kappes-Becker
- Centro de Esportes, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Elisio Alberto Evangelista
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| | - Renata Guerra-Sá
- Laboratório de Bioquímica e Biologia Molecular, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brasil
| |
Collapse
|
142
|
Sturla L, Mannino E, Scarfì S, Bruzzone S, Magnone M, Sociali G, Booz V, Guida L, Vigliarolo T, Fresia C, Emionite L, Buschiazzo A, Marini C, Sambuceti G, De Flora A, Zocchi E. Abscisic acid enhances glucose disposal and induces brown fat activity in adipocytes in vitro and in vivo. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:131-144. [PMID: 27871880 DOI: 10.1016/j.bbalip.2016.11.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/20/2016] [Accepted: 11/14/2016] [Indexed: 11/30/2022]
Abstract
Abscisic acid (ABA) is a plant hormone also present in animals, where it is involved in the regulation of innate immune cell function and of glucose disposal, through its receptor LANCL2. ABA stimulates glucose uptake by myocytes and pre-adipocytes in vitro and oral ABA improves glycemic control in rats and in healthy subjects. Here we investigated the role of the ABA/LANCL2 system in the regulation of glucose uptake and metabolism in adipocytes. Silencing of LANCL2 abrogated both the ABA- and insulin-induced increase of glucose transporter-4 expression and of glucose uptake in differentiated 3T3-L1 murine adipocytes; conversely, overexpression of LANCL2 enhanced basal, ABA- and insulin-stimulated glucose uptake. As compared with insulin, ABA treatment of adipocytes induced lower triglyceride accumulation, CO2 production and glucose-derived fatty acid synthesis. ABA per se did not induce pre-adipocyte differentiation in vitro, but stimulated adipocyte remodeling in terminally differentiated cells, with a reduction in cell size, increased mitochondrial content, enhanced O2 consumption, increased transcription of adiponectin and of brown adipose tissue (BAT) genes. A single dose of oral ABA (1μg/kg body weight) increased BAT glucose uptake 2-fold in treated rats compared with untreated controls. One-month-long ABA treatment at the same daily dose significantly upregulated expression of BAT markers in the WAT and in WAT-derived preadipocytes from treated mice compared with untreated controls. These results indicate a hitherto unknown role of LANCL2 in adipocyte sensitivity to insulin-stimulated glucose uptake and suggest a role for ABA in the induction and maintenance of BAT activity.
Collapse
Affiliation(s)
- Laura Sturla
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy.
| | - Elena Mannino
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Sonia Scarfì
- Department of Earth, Environment and Life Sciences, University of Genova, Via Pastore 3, 16132 Genova, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Mirko Magnone
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Giovanna Sociali
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Valeria Booz
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Lucrezia Guida
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Tiziana Vigliarolo
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Chiara Fresia
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Laura Emionite
- Animal Facility, IRCCS AOU San Martino - IST, Genova, Italy
| | - Ambra Buschiazzo
- Nuclear Medicine, Dept of Health Sciences, University of Genova, Genova, Italy
| | - Cecilia Marini
- CNR Institute of Bioimages and Molecular Physiology, Section of Genova, Genova, Italy; IRCCS AOU San Martino - IST, Genova, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine, Dept of Health Sciences, University of Genova, Genova, Italy; IRCCS AOU San Martino - IST, Genova, Italy
| | - Antonio De Flora
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| | - Elena Zocchi
- Department of Experimental Medicine and CEBR, University of Genova, Viale Benedetto XV 1, 16132 Genova, Italy
| |
Collapse
|
143
|
Strycharz J, Drzewoski J, Szemraj J, Sliwinska A. Is p53 Involved in Tissue-Specific Insulin Resistance Formation? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9270549. [PMID: 28194257 PMCID: PMC5282448 DOI: 10.1155/2017/9270549] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/19/2016] [Indexed: 02/06/2023]
Abstract
p53 constitutes an extremely versatile molecule, primarily involved in sensing the variety of cellular stresses. Functional p53 utilizes a plethora of mechanisms to protect cell from deleterious repercussions of genotoxic insults, where senescence deserves special attention. While the impressive amount of p53 roles has been perceived solely by the prism of antioncogenic effect, its presence seems to be vastly connected with metabolic abnormalities underlain by cellular aging, obesity, and inflammation. p53 has been found to regulate multiple biochemical processes such as glycolysis, oxidative phosphorylation, lipolysis, lipogenesis, β-oxidation, gluconeogenesis, and glycogen synthesis. Notably, p53-mediated metabolic effects are totally up to results of insulin action. Accumulating amount of data identifies p53 to be a factor activated upon hyperglycemia or excessive calorie intake, thus contributing to low-grade chronic inflammation and systemic insulin resistance. Prominent signs of its actions have been observed in muscles, liver, pancreas, and adipose tissue being associated with attenuation of insulin signalling. p53 is of crucial importance for the regulation of white and brown adipogenesis simultaneously being a repressor for preadipocyte differentiation. This review provides a profound insight into p53-dependent metabolic actions directed towards promotion of insulin resistance as well as presenting experimental data regarding obesity-induced p53-mediated metabolic abnormalities.
Collapse
Affiliation(s)
- Justyna Strycharz
- Diabetes Student Scientific Society at the Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, Lodz, Poland
| | - Jozef Drzewoski
- Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Sliwinska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
144
|
Esteves JV, Enguita FJ, Machado UF. MicroRNAs-Mediated Regulation of Skeletal Muscle GLUT4 Expression and Translocation in Insulin Resistance. J Diabetes Res 2017; 2017:7267910. [PMID: 28428964 PMCID: PMC5385897 DOI: 10.1155/2017/7267910] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 01/12/2023] Open
Abstract
The solute carrier family 2 facilitated glucose transporter member 4 (GLUT4) plays a key role in the insulin-induced glucose uptake by muscle and adipose tissues. In prediabetes and diabetes, GLUT4 expression/translocation has been detected as reduced, participating in mechanisms that impair glycemic control. Recently, a class of short endogenous noncoding RNAs named microRNAs (miRNAs) has been increasingly described as involved in the posttranscriptional epigenetic regulation of gene expression. The present review focuses on miRNAs potentially involved in the expression of GLUT4 expression, and proteins related to GLUT4 and translocation in skeletal muscle, seeking to correlate them with insulin resistance and diabetes. So far, miR-21a-5p, miR-29a-3p, miR-29c-3p, miR-93-5p, miR-106b-5p, miR-133a-3p, miR-133b-3p, miR-222-3p, and miR-223-3p have been reported to directly and/or indirectly regulate the GLUT4 expression; and their expression is altered under diabetes-related conditions. Besides, some miRNAs that have been linked to the expression of proteins involved in GLUT4 translocation machinery in muscle could also impact glucose uptake. That makes these miRNAs promising targets for preventive and/or therapeutic approaches, which could improve glycemic control, thus deserving future new investigations.
Collapse
Affiliation(s)
- João Victor Esteves
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Francisco Javier Enguita
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ubiratan Fabres Machado
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
- *Ubiratan Fabres Machado:
| |
Collapse
|
145
|
Oh KJ, Lee DS, Kim WK, Han BS, Lee SC, Bae KH. Metabolic Adaptation in Obesity and Type II Diabetes: Myokines, Adipokines and Hepatokines. Int J Mol Sci 2016; 18:ijms18010008. [PMID: 28025491 PMCID: PMC5297643 DOI: 10.3390/ijms18010008] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/24/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Obesity and type II diabetes are characterized by insulin resistance in peripheral tissues. A high caloric intake combined with a sedentary lifestyle is the leading cause of these conditions. Whole-body insulin resistance and its improvement are the result of the combined actions of each insulin-sensitive organ. Among the fundamental molecular mechanisms by which each organ is able to communicate and engage in cross-talk are cytokines or peptides which stem from secretory organs. Recently, it was reported that several cytokines or peptides are secreted from muscle (myokines), adipose tissue (adipokines) and liver (hepatokines) in response to certain nutrition and/or physical activity conditions. Cytokines exert autocrine, paracrine or endocrine effects for the maintenance of energy homeostasis. The present review is focused on the relationship and cross-talk amongst muscle, adipose tissue and the liver as secretory organs in metabolic diseases.
Collapse
Affiliation(s)
- Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Da Som Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Baek Soo Han
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34141, Korea.
| |
Collapse
|
146
|
Shin J, Yang SJ, Lim Y. Gamma-tocopherol supplementation ameliorated hyper-inflammatory response during the early cutaneous wound healing in alloxan-induced diabetic mice. Exp Biol Med (Maywood) 2016; 242:505-515. [PMID: 28211759 DOI: 10.1177/1535370216683836] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Delayed wound healing is one of the major diabetic complications. During wound healing process, the early inflammatory stage is important for better prognosis. One of antioxidant nutrient, gamma-tocopherol (GT) is considered to regulate inflammatory conditions. This study investigated the effect of GT supplementation on mechanism associated with inflammation, oxidative stress, and apoptosis during early cutaneous wound healing in diabetic mice. Diabetes was induced by alloxan injection in ICR mice. All mice were divided into three groups: non-diabetic control mice (CON), diabetic control mice (DMC), and diabetic mice supplemented with GT (GT). After two weeks of GT supplementation, excisional wounds were made by biopsy punches (4 mm). Diabetic mice showed increases in fasting blood glucose (FBG) level, hyper-inflammatory response, oxidative stress, and delayed wound closure rate compared to non-diabetic mice. However, GT supplementation reduced FBG level and accelerated wound closure rate by regulation of inflammatory response-related proteins such as nuclear factor kappa B, interleukin-1β, tumor necrosis factor-α, and c-reactive protein, and oxidative stress-related markers including nuclear factor (erythroid derived 2)-like 2, NAD(P)H dehydrogenase quinone1, heme oxygenase-1, manganese superoxide dismutase, catalase and glutathione peroxidase and apoptosis-related markers such as sirtuin-1, peroxisome proliferator-activated receptor gamma coactivator 1- α, and p53 in diabetic mice. Taken together, GT would be a potential therapeutic to prevent diabetes-induced delayed wound healing by regulation of inflammatory response, apoptosis, and oxidative stress. Impact statement Gamma tocopherol has shown ameliorative effect on diabetic wound healing by regulation of inflammation, oxidative stress, and apoptosis demonstrated by nuclear factor kappa B, nuclear factor (erythroid derived 2)-like 2, and sirtuin-1.
Collapse
Affiliation(s)
- Jihyun Shin
- 1 Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Korea
| | - Soo Jin Yang
- 2 Department of Food and Nutrition, Seoul Women's University, Seoul 01797, Korea
| | - Yunsook Lim
- 1 Department of Food and Nutrition, Kyung Hee University, Seoul 02447, Korea
| |
Collapse
|
147
|
Abstract
Insulin resistance is one of the defining features of type 2 diabetes and the metabolic syndrome and accompanies many other clinical conditions, ranging from obesity to lipodystrophy to glucocorticoid excess. Extraordinary efforts have gone into defining the mechanisms that underlie insulin resistance, with most attention focused on altered signalling as well as mitochondrial and endoplasmic reticulum stress. Here, nuclear mechanisms of insulin resistance, including transcriptional and epigenomic effects, will be discussed. Three levels of control involving transcription factors, transcriptional cofactors, and chromatin-modifying enzymes will be considered. Well-studied examples of the first include PPAR-γ in adipose tissue and the glucocorticoid receptor and FoxO1 in a variety of insulin-sensitive tissues. These proteins work in concert with cofactors such as PGC-1α and CRTC2, and chromatin-modifying enzymes including DNA methyltransferases and histone acetyltransferases, to regulate key genes that promote insulin-stimulated glucose uptake, gluconeogenesis or other pathways that affect systemic insulin action. Furthermore, genetic variation associated with increased risk of type 2 diabetes is often related to altered transcription factor binding, either by affecting the transcription factor itself, or more commonly by changing the binding affinity of a noncoding regulatory region. Finally, several avenues for therapeutic exploitation in the battle against metabolic disease will be discussed, including small-molecule inhibitors and activators of these factors and their related pathways.
Collapse
Affiliation(s)
- E D Rosen
- Division of Endocrinology and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
148
|
Epoxyeicosatrienoic Acid as Therapy for Diabetic and Ischemic Cardiomyopathy. Trends Pharmacol Sci 2016; 37:945-962. [DOI: 10.1016/j.tips.2016.08.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/12/2016] [Accepted: 08/17/2016] [Indexed: 12/19/2022]
|
149
|
Abstract
Hibernation is characterized by prolonged periods of inactivity with concomitantly low nutrient intake, conditions that would typically result in muscle atrophy combined with a loss of oxidative fibers. Yet, hibernators consistently emerge from winter with very little atrophy, frequently accompanied by a slight shift in fiber ratios to more oxidative fiber types. Preservation of muscle morphology is combined with down-regulation of glycolytic pathways and increased reliance on lipid metabolism instead. Furthermore, while rates of protein synthesis are reduced during hibernation, balance is maintained by correspondingly low rates of protein degradation. Proposed mechanisms include a number of signaling pathways and transcription factors that lead to increased oxidative fiber expression, enhanced protein synthesis and reduced protein degradation, ultimately resulting in minimal loss of skeletal muscle protein and oxidative capacity. The functional significance of these outcomes is maintenance of skeletal muscle strength and fatigue resistance, which enables hibernating animals to resume active behaviors such as predator avoidance, foraging and mating immediately following terminal arousal in the spring.
Collapse
Affiliation(s)
- Clark J Cotton
- Department of Biology, College of St Benedict/St John's University, Collegeville, MN 56321, USA
| |
Collapse
|
150
|
Oladimeji P, Cui H, Zhang C, Chen T. Regulation of PXR and CAR by protein-protein interaction and signaling crosstalk. Expert Opin Drug Metab Toxicol 2016; 12:997-1010. [PMID: 27295009 DOI: 10.1080/17425255.2016.1201069] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Protein-protein interaction and signaling crosstalk contribute to the regulation of pregnane X receptor (PXR) and constitutive androstane receptor (CAR) and broaden their cellular function. AREA COVERED This review covers key historic discoveries and recent advances in our understanding of the broad function of PXR and CAR and their regulation by protein-protein interaction and signaling crosstalk. EXPERT OPINION PXR and CAR were first discovered as xenobiotic receptors; however, it is clear that PXR and CAR perform a much broader range of cellular functions through protein-protein interaction and signaling crosstalk, which typically mutually affect the function of all the partners involved. Future research on PXR and CAR should, therefore, look beyond their xenobiotic function.
Collapse
Affiliation(s)
- Peter Oladimeji
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Hongmei Cui
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Chen Zhang
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| | - Taosheng Chen
- a Department of Chemical Biology and Therapeutics , St. Jude Children's Research Hospital , Memphis , TN , USA
| |
Collapse
|