1
|
Xu M, Lv D, Wei H, Li Z, Jin S, Liu Q, Zhang Y, Liu Y. Effects of antidiabetic agents on lipid metabolism of skeletal muscle: A narrative review. Diabetes Obes Metab 2025. [PMID: 39807619 DOI: 10.1111/dom.16189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/17/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025]
Abstract
Metabolic syndrome-related diseases frequently involve disturbances in skeletal muscle lipid metabolism. The accumulation of lipid metabolites, lipid-induced mitochondrial stress in skeletal muscle cells, as well as the inflammation of adjacent adipose tissue, are associated with the development of insulin resistance and metabolic dysfunction. Consequently, when antidiabetic medications are used to treat various chronic conditions related to hyperglycaemia, the impact on skeletal muscle lipid metabolism should not be overlooked. However, current research has predominantly focused on muscle mass rather than skeletal muscle lipid metabolism and its interplay with glucose metabolism. In this review, we summarised the latest research on the effects of antidiabetic drugs and certain natural compounds with antidiabetic activity on skeletal muscle lipid metabolism, focusing on data from preclinical to clinical studies. Given the widespread use of antidiabetic drugs, a better understanding of their effects on skeletal muscle lipid metabolism merits further attention in future research.
Collapse
Affiliation(s)
- Ming Xu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Dongqing Lv
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Hongxia Wei
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhe Li
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Shuqing Jin
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Qinhao Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
2
|
Li Y, Pan Y, Zhao X, Wu S, Li F, Wang Y, Liu B, Zhang Y, Gao X, Wang Y, Zhou H. Peroxisome proliferator-activated receptors: A key link between lipid metabolism and cancer progression. Clin Nutr 2024; 43:332-345. [PMID: 38142478 DOI: 10.1016/j.clnu.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023]
Abstract
Lipids represent the essential components of membranes, serve as fuels for high-energy processes, and play crucial roles in signaling and cellular function. One of the key hallmarks of cancer is the reprogramming of metabolic pathways, especially abnormal lipid metabolism. Alterations in lipid uptake, lipid desaturation, de novo lipogenesis, lipid droplets, and fatty acid oxidation in cancer cells all contribute to cell survival in a changing microenvironment by regulating feedforward oncogenic signals, key oncogenic functions, oxidative and other stresses, immune responses, or intercellular communication. Peroxisome proliferator-activated receptors (PPARs) are transcription factors activated by fatty acids and act as core lipid sensors involved in the regulation of lipid homeostasis and cell fate. In addition to regulating whole-body energy homeostasis in physiological states, PPARs play a key role in lipid metabolism in cancer, which is receiving increasing research attention, especially the fundamental molecular mechanisms and cancer therapies targeting PPARs. In this review, we discuss how cancer cells alter metabolic patterns and regulate lipid metabolism to promote their own survival and progression through PPARs. Finally, we discuss potential therapeutic strategies for targeting PPARs in cancer based on recent studies from the last five years.
Collapse
Affiliation(s)
- Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yujie Pan
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yuxiong Wang
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanghe Zhang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Xin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
3
|
Arslan S, Yıldıran H, Seymen CM. The Effect of Maternal High-Fat Diet on Adipose Tissue Histology and Lipid Metabolism-Related Genes Expression in Offspring Rats. Nutrients 2024; 16:150. [PMID: 38201978 PMCID: PMC10780511 DOI: 10.3390/nu16010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The developing fetus is dependent on the maternal nutritional environment. This study was conducted to determine the effects of a maternal high-fat diet (HFD) applied during pregnancy and/or lactation on the expression levels of some lipid-related genes in rat models. Half of the pregnant rats (n: 6) were fed an HFD (energy from fat: 45%), while the other half (n: 6) were fed a control diet (CD) (energy from fat, 7.7%) during the pregnancy period. During lactation, dams in both groups were divided into two subgroups, with half fed the CD and the other half fed the HFD. Thus, four groups were obtained: CD-CD, CD-HFD, HFD-CD, and HFD-HFD. At the end of lactation, all mothers and half of the offspring were sacrificed. The remaining offspring were fed a CD for five weeks. The average birth weight of the CD group offspring was found to be lower than that of the HFD group (p < 0.05). The amount of adipose tissue was highest in CD-HFD (p < 0.05), while gene expression levels were similar between groups (p > 0.05), and the most degenerative histological changes were observed in the eight-week HFD-HFD (p < 0.05). This study suggests that maternal HFD during pregnancy and lactation may increase adiposity in offspring rats, especially during the weaning period.
Collapse
Affiliation(s)
- Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Hilal Yıldıran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Turkey;
| | - Cemile Merve Seymen
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06500, Turkey;
| |
Collapse
|
4
|
Hill DP, Drabkin HJ, Smith CL, Van Auken KM, D’Eustachio P. Biochemical pathways represented by Gene Ontology-Causal Activity Models identify distinct phenotypes resulting from mutations in pathways. Genetics 2023; 225:iyad152. [PMID: 37579192 PMCID: PMC10550311 DOI: 10.1093/genetics/iyad152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/13/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023] Open
Abstract
Gene inactivation can affect the process(es) in which that gene acts and causally downstream ones, yielding diverse mutant phenotypes. Identifying the genetic pathways resulting in a given phenotype helps us understand how individual genes interact in a functional network. Computable representations of biological pathways include detailed process descriptions in the Reactome Knowledgebase and causal activity flows between molecular functions in Gene Ontology-Causal Activity Models (GO-CAMs). A computational process has been developed to convert Reactome pathways to GO-CAMs. Laboratory mice are widely used models of normal and pathological human processes. We have converted human Reactome GO-CAMs to orthologous mouse GO-CAMs, as a resource to transfer pathway knowledge between humans and model organisms. These mouse GO-CAMs allowed us to define sets of genes that function in a causally connected way. To demonstrate that individual variant genes from connected pathways result in similar but distinguishable phenotypes, we used the genes in our pathway models to cross-query mouse phenotype annotations in the Mouse Genome Database (MGD). Using GO-CAM representations of 2 related but distinct pathways, gluconeogenesis and glycolysis, we show that individual causal paths in gene networks give rise to discrete phenotypic outcomes resulting from perturbations of glycolytic and gluconeogenic genes. The accurate and detailed descriptions of gene interactions recovered in this analysis of well-studied processes suggest that this strategy can be applied to less well-understood processes in less well-studied model systems to predict phenotypic outcomes of novel gene variants and to identify potential gene targets in altered processes.
Collapse
Affiliation(s)
- David P Hill
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | | | - Kimberly M Van Auken
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Peter D’Eustachio
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
5
|
Miao M, Wang X, Liu T, Li YJ, Yu WQ, Yang TM, Guo SD. Targeting PPARs for therapy of atherosclerosis: A review. Int J Biol Macromol 2023:125008. [PMID: 37217063 DOI: 10.1016/j.ijbiomac.2023.125008] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
Atherosclerosis, a chief pathogenic factor of cardiovascular disease, is associated with many factors including inflammation, dyslipidemia, and oxidative stress. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors and are widely expressed with tissue- and cell-specificity. They control multiple genes that are involved in lipid metabolism, inflammatory response, and redox homeostasis. Given the diverse biological functions of PPARs, they have been extensively studied since their discovery in 1990s. Although controversies exist, accumulating evidence have demonstrated that PPAR activation attenuates atherosclerosis. Recent advances are valuable for understanding the mechanisms of action of PPAR activation. This article reviews the recent findings, mainly from the year of 2018 to present, including endogenous molecules in regulation of PPARs, roles of PPARs in atherosclerosis by focusing on lipid metabolism, inflammation, and oxidative stress, and synthesized PPAR modulators. This article provides information valuable for researchers in the field of basic cardiovascular research, for pharmacologists that are interested in developing novel PPAR agonists and antagonists with lower side effects as well as for clinicians.
Collapse
Affiliation(s)
- Miao Miao
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Xue Wang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Tian Liu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Yan-Jie Li
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Wen-Qian Yu
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Tong-Mei Yang
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Shou-Dong Guo
- Institute of Lipid Metabolism and Atherosclerosis, Innovative Drug Research Centre, School of Pharmacy, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
6
|
Melloni E, Grassilli S, Romani A, Rimondi E, Marcuzzi A, Zauli E, Secchiero P, Paganetto G, Guerrini A, Sacchetti G, Tacchini M. Convolvulus pluricaulis Choisy’s Extraction, Chemical Characterization and Evaluation of the Potential Effects on Glycaemic Balance in a 3T3-L1 Adipocyte Cell Model. Nutrients 2023; 15:nu15071727. [PMID: 37049568 PMCID: PMC10097163 DOI: 10.3390/nu15071727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Convolvulus pluricaulis (CP) is a common Indian herb, largely employed in Ayurvedic medicine and known for its neuroprotective and neuroinflammatory action. Its effectiveness against several pathologic/sub-pathologic conditions is widely accepted, but it is not yet completely chemically characterized. In recent years, several researchers have pointed out the involvement of CP and other Convolvulaceae in lipidic and glucidic metabolism, particularly in the control of hyperlipidaemia and diabetic conditions. In this scenario, the aim of the study was to chemically characterize the medium polarity part of the CP whole plant and its fractions and to shed light on their biological activity in adipocyte differentiation using the 3T3-L1 cell model. Our results demonstrated that the CP extract and fractions could upregulate the adipocyte differentiation through the modulation of the nuclear receptor PPARγ (Peroxisome Proliferator-Activated Receptor γ), broadly recognized as a key regulator of adipocyte differentiation, and the glucose transporter GLUT-4, which is fundamental for cellular glucose uptake and for metabolism control. CP also showed the ability to exert an anti-inflammatory effect, downregulating cytokines such as Rantes, MCP-1, KC, eotaxin, and GM-CSF, which are deeply involved in insulin resistance and glucose intolerance. Taken together, these data suggest that CP could exert a potential beneficial effect on glycemia and could be employed as an anti-diabetic adjuvant or, in any case, a means to better control glucose homeostasis.
Collapse
Affiliation(s)
- Elisabetta Melloni
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Silvia Grassilli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Arianna Romani
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Erika Rimondi
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Annalisa Marcuzzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Enrico Zauli
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Secchiero
- Department of Translational Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy
| | - Guglielmo Paganetto
- Department of Life Sciences and Biotechnology (SVeB), UR7 Terra&Acqua Tech, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandra Guerrini
- Department of Life Sciences and Biotechnology (SVeB), UR7 Terra&Acqua Tech, University of Ferrara, 44121 Ferrara, Italy
| | - Gianni Sacchetti
- Department of Life Sciences and Biotechnology (SVeB), UR7 Terra&Acqua Tech, University of Ferrara, 44121 Ferrara, Italy
| | - Massimo Tacchini
- Department of Life Sciences and Biotechnology (SVeB), UR7 Terra&Acqua Tech, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
7
|
Role of ACSL5 in fatty acid metabolism. Heliyon 2023; 9:e13316. [PMID: 36816310 PMCID: PMC9932481 DOI: 10.1016/j.heliyon.2023.e13316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/07/2022] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Free fatty acids (FFAs) are essential energy sources for most body tissues. A fatty acid must be converted to fatty acyl-CoA to oxidize or be incorporated into new lipids. Acyl-CoA synthetase long-chain family member 5 (ACSL5) is localized in the endoplasmic reticulum and mitochondrial outer membrane, where it catalyzes the formation of fatty acyl-CoAs from long-chain fatty acids (C16-C20). Fatty acyl-CoAs are then used in lipid synthesis or β-oxidation mediated pathways. ACSL5 plays a pleiotropic role in lipid metabolism depending on substrate preferences, subcellular localization and tissue specificity. Here, we review the role of ACSL5 in fatty acid metabolism in multiple metabolic tissues, including the liver, small intestine, adipose tissue, and skeletal muscle. Given the increasing number of studies suggesting the role of ACSL5 in glucose and lipid metabolism, we also summarized the effects of ACSL5 on circulating lipids and insulin resistance.
Collapse
|
8
|
Tranganida A, Hall AJ, Armstrong HC, Moss SEW, Bennett KA. Consequences of in vitro benzyl butyl phthalate exposure for blubber gene expression and insulin-induced Akt activation in juvenile grey seals. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 316:120688. [PMID: 36402420 DOI: 10.1016/j.envpol.2022.120688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/27/2022] [Accepted: 11/15/2022] [Indexed: 06/16/2023]
Abstract
Plastic and plasticiser pollution of marine environments is a growing concern. Although phthalates, one group of plasticisers, are rapidly metabolised by mammals, they are found ubiquitously in humans and have been linked with metabolic disorders and altered adipose function. Phthalates may also present a threat to marine mammals, which need to rapidly accumulate and mobilise their large fat depots. High molecular weight (HMW) phthalates may be most problematic because they can accumulate in adipose. We used blubber explants from juvenile grey seals to examine the effects of overnight exposure to the HMW, adipogenic phthalate, benzyl butyl phthalate (BBzP) on expression of key adipose-specific genes and on phosphorylation of Akt in response to insulin. We found substantial differences in transcript abundance of Pparγ, Insig2, Fasn, Scd, Adipoq and Lep between moult stages, when animals were also experiencing differing mass changes, and between tissue depths, which likely reflect differences in blubber function. Akt abundance was higher in inner compared to outer blubber, consistent with greater metabolic activity in adipose closer to muscle than skin, and its phosphorylation was stimulated by insulin. Transcript abundance of Pparγ and Fasn (and Adipoq in some animals) were increased by short term (30 min) insulin exposure. In addition, overnight in vitro BBzP exposure altered insulin-induced changes in Pparγ (and Adipoq in some animals) transcript abundance, in a tissue depth and moult stage-specific manner. Basal or insulin-induced Akt phosphorylation was not changed. BBzP thus acted rapidly on the transcript abundance of key adipose genes in an Akt-independent manner. Our data suggest phthalate exposure could alter seal blubber development or function, although the whole animal consequences of these changes are not yet understood. Knowledge of typical phthalate exposures and toxicokinetics would help to contextualise these findings in terms of phthalate-induced metabolic disruption risk and consequences for marine mammal health.
Collapse
Affiliation(s)
- Alexandra Tranganida
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, KY16 8LB, UK; Division of Health Science, School of Applied Sciences, Abertay University, Dundee, DD1 1HG, UK
| | - Ailsa J Hall
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, KY16 8LB, UK
| | - Holly C Armstrong
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, KY16 8LB, UK; Division of Health Science, School of Applied Sciences, Abertay University, Dundee, DD1 1HG, UK; School of Psychology and Neuroscience, University of St Andrews, KY16 9JP, UK
| | - Simon E W Moss
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St Andrews, KY16 8LB, UK
| | - Kimberley A Bennett
- Division of Health Science, School of Applied Sciences, Abertay University, Dundee, DD1 1HG, UK.
| |
Collapse
|
9
|
Marketou M, Lazopoulos G, Kontaraki J, Kalogerakos P, Plevritaki A, Chlouverakis G, Fragiadakis K, Maragkoudakis S, Zervakis S, Savva E, Kampanieris E, Kochiadakis G. PPAR-γ gene expression in pericoronary adipose tissue: A focus on obesity. Hellenic J Cardiol 2023; 69:67-68. [PMID: 36179807 DOI: 10.1016/j.hjc.2022.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/22/2022] [Accepted: 09/25/2022] [Indexed: 02/07/2023] Open
Affiliation(s)
- Maria Marketou
- Cardiology Department, Heraklion University General Hospital, Crete, Greece.
| | - George Lazopoulos
- Cardiovascular Surgery Department, Heraklion University General Hospital, Crete, Greece
| | - Joanna Kontaraki
- Molecular Cardiology Laboratory, School of Medicine, University of Crete, Crete, Greece
| | - Paris Kalogerakos
- Cardiovascular Surgery Department, Heraklion University General Hospital, Crete, Greece
| | | | - Gregory Chlouverakis
- Department of Biostatistics, School of Medicine, University of Crete, Crete, Greece
| | | | | | - Stelios Zervakis
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| | - Eirini Savva
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| | | | - George Kochiadakis
- Cardiology Department, Heraklion University General Hospital, Crete, Greece
| |
Collapse
|
10
|
Anwar MY, Baldassari AR, Polikowsky HG, Sitlani CM, Highland HM, Chami N, Chen HH, Graff M, Howard AG, Jung SY, Petty LE, Wang Z, Zhu W, Buyske S, Cheng I, Kaplan R, Kooperberg C, Loos RJF, Peters U, McCormick JB, Fisher-Hoch SP, Avery CL, Taylor KC, Below JE, North KE. Genetic pleiotropy underpinning adiposity and inflammation in self-identified Hispanic/Latino populations. BMC Med Genomics 2022; 15:192. [PMID: 36088317 PMCID: PMC9464371 DOI: 10.1186/s12920-022-01352-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/02/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Concurrent variation in adiposity and inflammation suggests potential shared functional pathways and pleiotropic disease underpinning. Yet, exploration of pleiotropy in the context of adiposity-inflammation has been scarce, and none has included self-identified Hispanic/Latino populations. Given the high level of ancestral diversity in Hispanic American population, genetic studies may reveal variants that are infrequent/monomorphic in more homogeneous populations. METHODS Using multi-trait Adaptive Sum of Powered Score (aSPU) method, we examined individual and shared genetic effects underlying inflammatory (CRP) and adiposity-related traits (Body Mass Index [BMI]), and central adiposity (Waist to Hip Ratio [WHR]) in HLA participating in the Population Architecture Using Genomics and Epidemiology (PAGE) cohort (N = 35,871) with replication of effects in the Cameron County Hispanic Cohort (CCHC) which consists of Mexican American individuals. RESULTS Of the > 16 million SNPs tested, variants representing 7 independent loci were found to illustrate significant association with multiple traits. Two out of 7 variants were replicated at statistically significant level in multi-trait analyses in CCHC. The lead variant on APOE (rs439401) and rs11208712 were found to harbor multi-trait associations with adiposity and inflammation. CONCLUSIONS Results from this study demonstrate the importance of considering pleiotropy for improving our understanding of the etiology of the various metabolic pathways that regulate cardiovascular disease development.
Collapse
Affiliation(s)
- Mohammad Yaser Anwar
- Department of Epidemiology, University of North Carolina at Chapel Hill, 123 West Franklin Street, CVD Genetic Epidemiology Lab, Fl #4, Room A7, Chapel Hill, NC, 27599, USA.
| | - Antoine R Baldassari
- Department of Epidemiology, University of North Carolina at Chapel Hill, 123 West Franklin Street, CVD Genetic Epidemiology Lab, Fl #4, Room A7, Chapel Hill, NC, 27599, USA
| | - Hannah G Polikowsky
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Colleen M Sitlani
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Heather M Highland
- Department of Epidemiology, University of North Carolina at Chapel Hill, 123 West Franklin Street, CVD Genetic Epidemiology Lab, Fl #4, Room A7, Chapel Hill, NC, 27599, USA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Mariaelisa Graff
- Department of Epidemiology, University of North Carolina at Chapel Hill, 123 West Franklin Street, CVD Genetic Epidemiology Lab, Fl #4, Room A7, Chapel Hill, NC, 27599, USA
| | - Annie Green Howard
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27516, USA
| | - Su Yon Jung
- Translational Sciences Section, School of Nursing, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Lauren E Petty
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wanying Zhu
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Steven Buyske
- Department of Statistics, Rutgers University, Piscataway, NJ, 08854, USA
| | - Iona Cheng
- Department of Epidemiology and Biostatistics, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Robert Kaplan
- Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Joseph B McCormick
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, 78520, USA
| | - Susan P Fisher-Hoch
- School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, 78520, USA
| | - Christy L Avery
- Department of Epidemiology, University of North Carolina at Chapel Hill, 123 West Franklin Street, CVD Genetic Epidemiology Lab, Fl #4, Room A7, Chapel Hill, NC, 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27516, USA
| | - Kira C Taylor
- Department of Epidemiology and Population Health, University of Louisville School of Public Health and Information Sciences, Louisville, KT, 40202, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina at Chapel Hill, 123 West Franklin Street, CVD Genetic Epidemiology Lab, Fl #4, Room A7, Chapel Hill, NC, 27599, USA
| |
Collapse
|
11
|
Park SS, Perez Perez JL, Perez Gandara B, Agudelo CW, Rodriguez Ortega R, Ahmed H, Garcia-Arcos I, McCarthy C, Geraghty P. Mechanisms Linking COPD to Type 1 and 2 Diabetes Mellitus: Is There a Relationship between Diabetes and COPD? Medicina (B Aires) 2022; 58:medicina58081030. [PMID: 36013497 PMCID: PMC9415273 DOI: 10.3390/medicina58081030] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 01/09/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) patients frequently suffer from multiple comorbidities, resulting in poor outcomes for these patients. Diabetes is observed at a higher frequency in COPD patients than in the general population. Both type 1 and 2 diabetes mellitus are associated with pulmonary complications, and similar therapeutic strategies are proposed to treat these conditions. Epidemiological studies and disease models have increased our knowledge of these clinical associations. Several recent genome-wide association studies have identified positive genetic correlations between lung function and obesity, possibly due to alterations in genes linked to cell proliferation; embryo, skeletal, and tissue development; and regulation of gene expression. These studies suggest that genetic predisposition, in addition to weight gain, can influence lung function. Cigarette smoke exposure can also influence the differential methylation of CpG sites in genes linked to diabetes and COPD, and smoke-related single nucleotide polymorphisms are associated with resting heart rate and coronary artery disease. Despite the vast literature on clinical disease association, little direct mechanistic evidence is currently available demonstrating that either disease influences the progression of the other, but common pharmacological approaches could slow the progression of these diseases. Here, we review the clinical and scientific literature to discuss whether mechanisms beyond preexisting conditions, lifestyle, and weight gain contribute to the development of COPD associated with diabetes. Specifically, we outline environmental and genetic confounders linked with these diseases.
Collapse
Affiliation(s)
- Sangmi S. Park
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Jessica L. Perez Perez
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Brais Perez Gandara
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Christina W. Agudelo
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Romy Rodriguez Ortega
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Huma Ahmed
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Itsaso Garcia-Arcos
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
| | - Cormac McCarthy
- University College Dublin School of Medicine, Education and Research Centre, St. Vincent’s University Hospital, D04 T6F4 Dublin, Ireland;
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA; (S.S.P.); (J.L.P.P.); (B.P.G.); (C.W.A.); (R.R.O.); (H.A.); (I.G.-A.)
- Correspondence: ; Tel.: +1-718-270-3141
| |
Collapse
|
12
|
Mirzaei-Alamouti H, Elhami S, Abdollahi A, Vazirigohar M, Harakinejad T, Nielson MO, Aschenbach JR, Mansouryar M. Short communication: effect of dietary supplementation with a mixture of fish and sunflower oils on the expression of key lipogenic and cholesterologenic genes in adipose tissues with different metabolic functions. Trop Anim Health Prod 2021; 53:522. [PMID: 34697645 DOI: 10.1007/s11250-021-02972-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/14/2021] [Indexed: 10/20/2022]
Abstract
The present study investigated the effects of dietary fish and sunflower oils as sources of n-3, n-6 polyunsaturated fatty acids (PUFA) on the expression of key lipogenic and cholesterologenic genes in subcutaneous adipose tissue (SAT) and tail adipose tissue (TAT) of fat-tailed sheep. Twenty-six male Afshari lambs were divided into 4 groups. Three groups were fed a high concentrate basal diet plus 100 g/lamb/day oil supplement (OS; 60 g sunflower oil and 40 g fish oil) beyond a 21-day adaptation period for 10, 20, and 30 days (groups OS10, OS20, and OS30; n = 6, each) until slaughter. A control group was slaughtered at the last day of adaptation (OS0; n = 4). Expression of PPARγ, SREBP-1c, and SREBP-2 were determined in TAT and SAT. All transcription factors had lower expression in SAT than TAT. Feeding OS induced a similar pattern of SREBP-1c expression in both TAT and SAT with highest values in OS20. SREBP-2 mRNA decreased by > 50% in TAT of OS30 compared to OS0, whereas the expression of SREBP-2 mRNA did not change in SAT in the same period. PPARγ expression was not affected over time either in SAT or TAT. Plasma concentrations of cholesterol and blood urea nitrogen increased in OS20. The comparison of gene expression responses to OS in TAT vs. SAT suggest that PUFA-mediated effects on lipid metabolism differ between SAT and TAT, which may be linked to the specific role of TAT in energy and water balance under arid conditions.
Collapse
Affiliation(s)
- H Mirzaei-Alamouti
- Department of Animal Sciences, Faculty of Agriculture, University of Zanjan, 45371-38791, Zanjan, Iran.
| | - S Elhami
- Department of Animal Sciences, Faculty of Agriculture, University of Zanjan, 45371-38791, Zanjan, Iran
| | - A Abdollahi
- Department of Animal Science, Faculty of Agriculture, Shiraz University, 71441-65186, Shiraz, Iran
| | - M Vazirigohar
- Zist Dam Group, University of Zanjan Incubator Center, 45371-38791, Zanjan, Iran
| | - T Harakinejad
- Department of Animal Sciences, Faculty of Agriculture, University of Zanjan, 45371-38791, Zanjan, Iran
| | - M O Nielson
- Department of Animal Science, Faculty of Science and Technology, Aarhus University, Blichers Allé, 20, 8830, Tjele, Denmark
| | - J R Aschenbach
- Institute of Veterinary Physiology, Freie Universität Berlin, Oertzenweg 19b, 14163, Berlin, Germany
| | - M Mansouryar
- Zist Dam Group, University of Zanjan Incubator Center, 45371-38791, Zanjan, Iran.
| |
Collapse
|
13
|
Saunders AM, Burns DK, Gottschalk WK. Reassessment of Pioglitazone for Alzheimer's Disease. Front Neurosci 2021; 15:666958. [PMID: 34220427 PMCID: PMC8243371 DOI: 10.3389/fnins.2021.666958] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is a quintessential 'unmet medical need', accounting for ∼65% of progressive cognitive impairment among the elderly, and 700,000 deaths in the United States in 2020. In 2019, the cost of caring for Alzheimer's sufferers was $244B, not including the emotional and physical toll on caregivers. In spite of this dismal reality, no treatments are available that reduce the risk of developing AD or that offer prolonged mitiagation of its most devestating symptoms. This review summarizes key aspects of the biology and genetics of Alzheimer's disease, and we describe how pioglitazone improves many of the patholophysiological determinants of AD. We also summarize the results of pre-clinical experiments, longitudinal observational studies, and clinical trials. The results of animal testing suggest that pioglitazone can be corrective as well as protective, and that its efficacy is enhanced in a time- and dose-dependent manner, but the dose-effect relations are not monotonic or sigmoid. Longitudinal cohort studies suggests that it delays the onset of dementia in individuals with pre-existing type 2 diabetes mellitus, which small scale, unblinded pilot studies seem to confirm. However, the results of placebo-controlled, blinded clinical trials have not borne this out, and we discuss possible explanations for these discrepancies.
Collapse
Affiliation(s)
- Ann M. Saunders
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | - Daniel K. Burns
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | | |
Collapse
|
14
|
Wagner G, Fenzl A, Lindroos-Christensen J, Einwallner E, Husa J, Witzeneder N, Rauscher S, Gröger M, Derdak S, Mohr T, Sutterlüty H, Klinglmüller F, Wolkerstorfer S, Fondi M, Hoermann G, Cao L, Wagner O, Kiefer FW, Esterbauer H, Bilban M. LMO3 reprograms visceral adipocyte metabolism during obesity. J Mol Med (Berl) 2021; 99:1151-1171. [PMID: 34018016 PMCID: PMC8313462 DOI: 10.1007/s00109-021-02089-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023]
Abstract
Abstract Obesity and body fat distribution are important risk factors for the development of type 2 diabetes and metabolic syndrome. Evidence has accumulated that this risk is related to intrinsic differences in behavior of adipocytes in different fat depots. We recently identified LIM domain only 3 (LMO3) in human mature visceral adipocytes; however, its function in these cells is currently unknown. The aim of this study was to determine the potential involvement of LMO3-dependent pathways in the modulation of key functions of mature adipocytes during obesity. Based on a recently engineered hybrid rAAV serotype Rec2 shown to efficiently transduce both brown adipose tissue (BAT) and white adipose tissue (WAT), we delivered YFP or Lmo3 to epididymal WAT (eWAT) of C57Bl6/J mice on a high-fat diet (HFD). The effects of eWAT transduction on metabolic parameters were evaluated 10 weeks later. To further define the role of LMO3 in insulin-stimulated glucose uptake, insulin signaling, adipocyte bioenergetics, as well as endocrine function, experiments were conducted in 3T3-L1 adipocytes and newly differentiated human primary mature adipocytes, engineered for transient gain or loss of LMO3 expression, respectively. AAV transduction of eWAT results in strong and stable Lmo3 expression specifically in the adipocyte fraction over a course of 10 weeks with HFD feeding. LMO3 expression in eWAT significantly improved insulin sensitivity and healthy visceral adipose tissue expansion in diet-induced obesity, paralleled by increased serum adiponectin. In vitro, LMO3 expression in 3T3-L1 adipocytes increased PPARγ transcriptional activity, insulin-stimulated GLUT4 translocation and glucose uptake, as well as mitochondrial oxidative capacity in addition to fatty acid oxidation. Mechanistically, LMO3 induced the PPARγ coregulator Ncoa1, which was required for LMO3 to enhance glucose uptake and mitochondrial oxidative gene expression. In human mature adipocytes, LMO3 overexpression promoted, while silencing of LMO3 suppressed mitochondrial oxidative capacity. LMO3 expression in visceral adipose tissue regulates multiple genes that preserve adipose tissue functionality during obesity, such as glucose metabolism, insulin sensitivity, mitochondrial function, and adiponectin secretion. Together with increased PPARγ activity and Ncoa1 expression, these gene expression changes promote insulin-induced GLUT4 translocation, glucose uptake in addition to increased mitochondrial oxidative capacity, limiting HFD-induced adipose dysfunction. These data add LMO3 as a novel regulator improving visceral adipose tissue function during obesity. Key messages LMO3 increases beneficial visceral adipose tissue expansion and insulin sensitivity in vivo. LMO3 increases glucose uptake and oxidative mitochondrial activity in adipocytes. LMO3 increases nuclear coactivator 1 (Ncoa1). LMO3-enhanced glucose uptake and mitochondrial gene expression requires Ncoa1.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-021-02089-9.
Collapse
Affiliation(s)
- Gabriel Wagner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Anna Fenzl
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090, Vienna, Austria
| | - Josefine Lindroos-Christensen
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,Novo Nordisk, Maaloev, Denmark
| | - Elisa Einwallner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Julia Husa
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Nadine Witzeneder
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Sabine Rauscher
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Marion Gröger
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Sophia Derdak
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Mohr
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Hedwig Sutterlüty
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Florian Klinglmüller
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, 1090, Vienna, Austria.,Austrian Medicines & Medical Devices Agency, 1200, Vienna, Austria
| | - Silviya Wolkerstorfer
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,University of Applied Sciences, FH Campus Wien, 1100, Vienna, Austria.,Institute of Cardiovascular Prevention, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Martina Fondi
- University of Applied Sciences, FH Campus Wien, 1100, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,Central Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Innsbruck, 6020, Innsbruck, Austria
| | - Lei Cao
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - Oswald Wagner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Florian W Kiefer
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090, Vienna, Austria
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria. .,Core Facilities, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
15
|
Kim SM, Imm JY. The Effect of Chrysin-Loaded Phytosomes on Insulin Resistance and Blood Sugar Control in Type 2 Diabetic db/db Mice. Molecules 2020; 25:molecules25235503. [PMID: 33255372 PMCID: PMC7727825 DOI: 10.3390/molecules25235503] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023] Open
Abstract
Although a variety of beneficial health effects of natural flavonoids, including chrysin, has been suggested, poor solubility and bioavailability limit their practical use. As a promising delivery system, chrysin-loaded phytosomes (CPs) were prepared using egg phospholipid (EPL) at a 1:3 molar ratio and its antidiabetic effects were assessed in db/db diabetic mice. Male C57BLKS/J-db/db mice were fed a normal diet (control), chrysin diet (100 mg chrysin/kg), CP diet (100 mg chrysin equivalent/kg), metformin diet (200 mg/kg) or EPL diet (vehicle, the same amount of EPL used for CP preparation) for 9 weeks. Administration of CP significantly decreased fasting blood glucose and insulin levels in db/db mice compared with the control. An oral glucose tolerance test and homeostatic model assessment for insulin resistance were significantly improved in the CP group (p < 0.05). CP treatment suppressed gluconeogenesis via downregulation of phosphoenolpyruvate carboxykinase while it promoted glucose uptake in the skeletal muscle and liver of db/db mice (p < 0.05). The CP-mediated improved glucose utilization in the muscle was confirmed by upregulation of glucose transporter type 4, hexokinase2 and peroxisome proliferator-activated receptor γ during treatment (p < 0.05). The CP-induced promotion of GLUT4 plasma translocation was confirmed in the skeletal muscle of db/db mice (p < 0.05). Based on the results, CP showed greater antidiabetic performance compared to the control by ameliorating insulin resistance in db/db mice and phytosome can be used as an effective antidiabetic agent.
Collapse
|
16
|
Singh R, Chandel S, Dey D, Ghosh A, Roy S, Ravichandiran V, Ghosh D. Epigenetic modification and therapeutic targets of diabetes mellitus. Biosci Rep 2020; 40:BSR20202160. [PMID: 32815547 PMCID: PMC7494983 DOI: 10.1042/bsr20202160] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
The prevalence of diabetes and its related complications are increasing significantly globally. Collected evidence suggested that several genetic and environmental factors contribute to diabetes mellitus. Associated complications such as retinopathy, neuropathy, nephropathy and other cardiovascular complications are a direct result of diabetes. Epigenetic factors include deoxyribonucleic acid (DNA) methylation and histone post-translational modifications. These factors are directly related with pathological factors such as oxidative stress, generation of inflammatory mediators and hyperglycemia. These result in altered gene expression and targets cells in the pathology of diabetes mellitus without specific changes in a DNA sequence. Environmental factors and malnutrition are equally responsible for epigenetic states. Accumulated evidence suggested that environmental stimuli alter the gene expression that result in epigenetic changes in chromatin. Recent studies proposed that epigenetics may include the occurrence of 'metabolic memory' found in animal studies. Further study into epigenetic mechanism might give us new vision into the pathogenesis of diabetes mellitus and related complication thus leading to the discovery of new therapeutic targets. In this review, we discuss the possible epigenetic changes and mechanism that happen in diabetes mellitus type 1 and type 2 separately. We highlight the important epigenetic and non-epigenetic therapeutic targets involved in the management of diabetes and associated complications.
Collapse
Affiliation(s)
- Rajveer Singh
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Shivani Chandel
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Dhritiman Dey
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Arijit Ghosh
- Department of Chemistry, University of Calcutta, Kolkata 700009, India
| | - Syamal Roy
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Velayutham Ravichandiran
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| | - Dipanjan Ghosh
- National Institute of Pharmaceutical Education and Research, Kolkata 164, Manicktala Main Road, Kolkata 700054, India
| |
Collapse
|
17
|
COVID-19: Proposing a Ketone-Based Metabolic Therapy as a Treatment to Blunt the Cytokine Storm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6401341. [PMID: 33014275 PMCID: PMC7519203 DOI: 10.1155/2020/6401341] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Human SARS-CoV-2 infection is characterized by a high mortality rate due to some patients developing a large innate immune response associated with a cytokine storm and acute respiratory distress syndrome (ARDS). This is characterized at the molecular level by decreased energy metabolism, altered redox state, oxidative damage, and cell death. Therapies that increase levels of (R)-beta-hydroxybutyrate (R-BHB), such as the ketogenic diet or consuming exogenous ketones, should restore altered energy metabolism and redox state. R-BHB activates anti-inflammatory GPR109A signaling and inhibits the NLRP3 inflammasome and histone deacetylases, while a ketogenic diet has been shown to protect mice from influenza virus infection through a protective γδ T cell response and by increasing electron transport chain gene expression to restore energy metabolism. During a virus-induced cytokine storm, metabolic flexibility is compromised due to increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) that damage, downregulate, or inactivate many enzymes of central metabolism including the pyruvate dehydrogenase complex (PDC). This leads to an energy and redox crisis that decreases B and T cell proliferation and results in increased cytokine production and cell death. It is hypothesized that a moderately high-fat diet together with exogenous ketone supplementation at the first signs of respiratory distress will increase mitochondrial metabolism by bypassing the block at PDC. R-BHB-mediated restoration of nucleotide coenzyme ratios and redox state should decrease ROS and RNS to blunt the innate immune response and the associated cytokine storm, allowing the proliferation of cells responsible for adaptive immunity. Limitations of the proposed therapy include the following: it is unknown if human immune and lung cell functions are enhanced by ketosis, the risk of ketoacidosis must be assessed prior to initiating treatment, and permissive dietary fat and carbohydrate levels for exogenous ketones to boost immune function are not yet established. The third limitation could be addressed by studies with influenza-infected mice. A clinical study is warranted where COVID-19 patients consume a permissive diet combined with ketone ester to raise blood ketone levels to 1 to 2 mM with measured outcomes of symptom severity, length of infection, and case fatality rate.
Collapse
|
18
|
Xie D, He Z, Dong Y, Gong Z, Nie G, Li Y. Molecular Cloning, Characterization, and Expression Regulation of Acyl-CoA Synthetase 6 Gene and Promoter in Common Carp Cyprinus carpio. Int J Mol Sci 2020; 21:E4736. [PMID: 32635148 PMCID: PMC7370118 DOI: 10.3390/ijms21134736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/27/2020] [Accepted: 06/28/2020] [Indexed: 02/08/2023] Open
Abstract
Omega-3 long chain polyunsaturated fatty acids (n-3 LC-PUFA), particularly docosahexaenoic acids (22:6n-3, DHA), have positive effects on multiple biologic and pathologic processes. Fish are the major dietary source of n-3 LC-PUFA for humans. Growing evidence supports acyl-coenzyme A (acyl-CoA) synthetase 6 (acsl6) being involved in cellular DHA uptake and lipogenesis in mammals, while its molecular function and regulatory mechanism remain unknown in fish. The present study focused on investigating the molecular characterization and transcription regulation of the acsl6 gene in the freshwater teleost common carp (Cyprinus carpio). First, the full length of acsl6 cDNA contained a coding region of 2148 bp for 715 amino acids, which possessed all characteristic features of the acyl-CoA synthetase (ACSL) family. Its mRNA expression was the highest in the brain, followed by in the heart, liver, kidney, muscle, and eyes, but little expression was detected in the ovary and gills. Additionally, a candidate acsl6 promoter region of 2058 bp was cloned, and the sequence from -758 bp to -198 bp was determined as core a promoter by equal progressive deletion and electrophoretic mobility shift assay. The binding sites for important transcription factors (TFs), including stimulatory protein 1 (SP1), CCAAT enhancer-binding protein (C/EBPα), sterol-regulatory element binding protein 1c (SREBP1c), peroxisome proliferator activated receptor α (PPARα), and PPARγ were identified in the core promoter by site-directed mutation and functional assays. Furthermore, the intraperitoneal injection of PPARγ agonists (balaglitazone) increased the expression of acsl6 mRNA, coupling with an increased proportion of DHA in the muscle, while opposite results were obtained in the injection of the SREBP1c antagonist (betulin). However, the expression of acsl6 and DHA content in muscle were largely unchanged by PPARα agonist (fenofibrate) treatment. These results indicated that acsl6 may play an important role for the muscular DHA uptake and deposition in common carp, and PPARγ and SREBP-1c are the potential TFs involved in the transcriptional regulation of acsl6 gene. To our knowledge, this is the first report of the characterization of acsl6 gene and its promoter in teleosts.
Collapse
Affiliation(s)
- Dizhi Xie
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; (D.X.); (Z.H.); (Y.D.)
| | - Zijie He
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; (D.X.); (Z.H.); (Y.D.)
- Laboratory of Aquatic Animal Nutrition and Diet, College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Yewei Dong
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; (D.X.); (Z.H.); (Y.D.)
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore 115473, Singapore;
| | - Guoxing Nie
- Laboratory of Aquatic Animal Nutrition and Diet, College of Fisheries, Henan Normal University, Xinxiang 453007, China
| | - Yuanyou Li
- College of Marine Sciences of South China Agricultural University & Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China; (D.X.); (Z.H.); (Y.D.)
| |
Collapse
|
19
|
Matsumoto Y, Ishimi Y, Suzuki T, Kobayashi KI, Inoue J, Yamamoto Y. Activation of peroxisome proliferator-activated receptor gamma/small heterodimer partner pathway prevents high fat diet-induced obesity and hepatic steatosis in Sprague-Dawley rats fed soybean meal. J Nutr Biochem 2020; 75:108250. [PMID: 31707284 DOI: 10.1016/j.jnutbio.2019.108250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 08/07/2019] [Accepted: 09/10/2019] [Indexed: 12/22/2022]
Abstract
Soybeans are a complete nutritional resource and soybean proteins are known to affect lipid metabolism via multiple mechanisms. Soybean meal (SBM) is produced after extraction of soybean oil and in this study, we investigated the ability whether the SBM could prevent high fat diet-induced obesity and understand the underlying mechanisms. Male Sprague-Dawley rats, aged 5 weeks, were randomly divided into three groups (n=8 each) and fed one of three diets for 28 days: Cont (AIN-93G), HFD (high fat diet with 40% of calories derived from fat) and HFD+SBM (HFD with 30% SBM). White adipose tissue weight as well as plasma and hepatic triglycerides were significantly decreased in HFD+SBM rats. Expression of hepatic SREBP-1 and its target genes was significantly decreased in HFD+SBM rats. Meanwhile, expression of SHP gene expression was significantly increased in HFD+SBM, and there was a negative correlation between SHP and SREBP-1 expression. Together these results suggest that hepatic SREBP-1 gene expression is negatively regulated by SHP. Expression of PPARG, the transcriptional factor that regulates SHP expression, was increased in HFD+SBM rats. Furthermore, expression of genes controlled by PPARG/SHP, such as those involved in hepatic gluconeogenesis, was also significantly decreased in HFD+SBM rats. Therefore, in addition to the previous findings of SBM on obesity here we show an additional mechanism which by changing the expression of genes involved in lipid metabolism via the PPARG/SHP pathway in the liver.
Collapse
Affiliation(s)
- Yu Matsumoto
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Yoshiko Ishimi
- Department of Food Function and Labeling, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8636, Japan
| | - Tsukasa Suzuki
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Ken-Ichi Kobayashi
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Jun Inoue
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan
| | - Yuji Yamamoto
- Department of Applied Biology and Chemistry, Faculty of Applied Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo 156-8502, Japan.
| |
Collapse
|
20
|
Hernandez-Olmos V, Knape T, Heering J, von Knethen A, Kilu W, Kaiser A, Wurglics M, Helmstädter M, Merk D, Schubert-Zsilavecz M, Parnham MJ, Steinhilber D, Proschak E. Structure optimization of a new class of PPARγ antagonists. Bioorg Med Chem 2019; 27:115082. [PMID: 31548084 DOI: 10.1016/j.bmc.2019.115082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) modulators have found wide application for the treatment of cancers, metabolic disorders and inflammatory diseases. Contrary to PPARγ agonists, PPARγ antagonists have been much less studied and although they have shown immunomodulatory effects, there is still no therapeutically useful PPARγ antagonist on the market. In contrast to non-competitive, irreversible inhibition caused by 2-chloro-5-nitrobenzanilide (GW9662), the recently described (E)-2-(5-((4-methoxy-2-(trifluoromethyl)quinolin-6-yl)methoxy)-2-((4-(trifluoromethyl)benzyl)oxy)-benzylidene)-hexanoic acid (MTTB, T-10017) is a promising prototype for a new class of PPARγ antagonists. It exhibits competitive antagonism against rosiglitazone mediated activation of PPARγ ligand binding domain (PPARγLBD) in a transactivation assay in HEK293T cells with an IC50 of 4.3 µM against 1 µM rosiglitazone. The aim of this study was to investigate the structure-activity relationships (SAR) of the MTTB scaffold focusing on improving its physicochemical properties. Through this optimization, 34 new derivatives were prepared and characterized. Two new potent compounds (T-10075 and T-10106) with much improved drug-like properties and promising pharmacokinetic profile were identified.
Collapse
Affiliation(s)
- Victor Hernandez-Olmos
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Tilo Knape
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Jan Heering
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Andreas von Knethen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Whitney Kilu
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Astrid Kaiser
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Mario Wurglics
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Moritz Helmstädter
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Manfred Schubert-Zsilavecz
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Dieter Steinhilber
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany
| | - Ewgenij Proschak
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology TMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
21
|
Woo MH, Lee HS, Kim J. Effect of pioglitazone in acute ischemic stroke patients with diabetes mellitus: a nested case-control study. Cardiovasc Diabetol 2019; 18:67. [PMID: 31151454 PMCID: PMC6545002 DOI: 10.1186/s12933-019-0874-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background Pioglitazone is an oral antidiabetic drug with multiple pleiotropic actions. Recent clinical trials have demonstrated that treatment with pioglitazone reduces cardiovascular risk in patients who have had an ischemic stroke. We examined the secondary preventive effects of pioglitazone in acute ischemic stroke patients with diabetes mellitus (DM) based on nationwide real-world data. Methods A nested case–control study was conducted with data from the National Health Insurance Service-National Sample Cohort in Korea. Study subjects were diabetic patients admitted for acute ischemic stroke (ICD-10 code; I63) between 2002 and 2013. Cases were defined as patients who suffered from composites of recurrent stroke (I60–63), myocardial infarction (I21), or all-cause mortality after ischemic stroke. Controls were selected by incidence density sampling. Three controls were matched to each case for sex, age, treatment with insulin, and oral antidiabetic medications, with the exception of pioglitazone. Medication history after ischemic stroke was obtained by accessing the prescription records. In the matched dataset, conditional logistic regression analysis was performed with adjustments for hypertension, atrial fibrillation, prior myocardial infarction, and treatment with oral antithrombotics and statins. Results From the patients with acute ischemic stroke and DM, 1150 cases with primary outcomes were matched to 3450 controls. In the matched analysis, treatment with pioglitazone was significantly associated with a lower cardiovascular risk (adjusted OR [95% CI], 0.43 [0.23–0.83]). Conclusions In this nested case–control study using real-world data, treatment with pioglitazone exhibited significant cardiovascular preventive effect in diabetic patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Min-Hee Woo
- Department of Neurology, CHA Bundang Medical Center, CHA University College of Medicine, Seongnam, Republic of Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinkwon Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University College of Medicine, Seongnam, Republic of Korea. .,Departments of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonjuro, Gangnam-gu, Seoul, 06273, Republic of Korea.
| |
Collapse
|
22
|
Vallée A, Vallée JN, Lecarpentier Y. Metabolic reprogramming in atherosclerosis: Opposed interplay between the canonical WNT/β-catenin pathway and PPARγ. J Mol Cell Cardiol 2019; 133:36-46. [PMID: 31153873 DOI: 10.1016/j.yjmcc.2019.05.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/27/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023]
Abstract
Atherosclerosis, a chronic inflammatory and age-related disease, is a complex mechanism presenting a dysregulation of vessel structures. During this process, the canonical WNT/β-catenin pathway is increased whereas PPARγ is downregulated. The two systems act in an opposite manner. This paper reviews the opposing interplay of these systems and their metabolic-reprogramming pathway in atherosclerosis. Activation of the WNT/β-catenin pathway enhances the transcription of targets involved in inflammation, endothelial dysfunction, the proliferation of vascular smooth muscle cells, and vascular calcification. This complex mechanism, which is partly controlled by the WNT/β-catenin pathway, presents several metabolic dysfunctions. This phenomenon, called aerobic glycolysis (or the Warburg effect), consists of a shift in ATP production from mitochondrial oxidative phosphorylation to aerobic glycolysis, leading to the overproduction of intracellular lactate. This mechanism is partially due to the injury of mitochondrial respiration and an increase in the glycolytic pathway. In contrast, PPARγ agonists downregulate the WNT/β-catenin pathway. Therefore, the development of therapeutic targets, such as PPARγ agonists, for the treatment of atherosclerosis could be an interesting and innovative way of counteracting the canonical WNT pathway.
Collapse
Affiliation(s)
- Alexandre Vallée
- Diagnosis and Therapeutic Center, Hypertension and Cardiovascular Prevention Unit, Hotel-Dieu Hospital, AP-HP, Université Paris Descartes, Paris, France.
| | - Jean-Noël Vallée
- Centre Hospitalier Universitaire (CHU) Amiens Picardie, Université Picardie Jules Verne (UPJV), 80054 Amiens, France; Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), 6-8 rue Saint-fiacre, 77100 Meaux, France
| |
Collapse
|
23
|
Shin SK, Cho HW, Song SE, Bae JH, Im SS, Hwang I, Ha H, Song DK. Ablation of catalase promotes non-alcoholic fatty liver via oxidative stress and mitochondrial dysfunction in diet-induced obese mice. Pflugers Arch 2019; 471:829-843. [PMID: 30617744 DOI: 10.1007/s00424-018-02250-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 12/05/2018] [Accepted: 12/26/2018] [Indexed: 12/12/2022]
Abstract
Hydrogen peroxide (H2O2) produced endogenously can cause mitochondrial dysfunction and metabolic complications in various cell types by inducing oxidative stress. In the liver, oxidative and endoplasmic reticulum (ER) stress affects the development of non-alcoholic fatty liver disease (NAFLD). Although a link between both stresses and fatty liver diseases has been suggested, few studies have investigated the involvement of catalase in fatty liver pathogenesis. We examined whether catalase is associated with NAFLD, using catalase knockout (CKO) mice and the catalase-deficient human hepatoma cell line HepG2. Hepatic morphology analysis revealed that the fat accumulation was more prominent in high-fat diet (HFD) CKO mice compared to that in age-matched wild-type (WT) mice, and lipid peroxidation and H2O2 release were significantly elevated in CKO mice. Transmission electron micrographs indicated that the liver mitochondria from CKO mice tended to be more severely damaged than those in WT mice. Likewise, mitochondrial DNA copy number and cellular ATP concentrations were significantly lower in CKO mice. In fatty acid-treated HepG2 cells, knockdown of catalase accelerated cellular lipid accumulation and depressed mitochondrial biogenesis, which was recovered by co-treatment with N-acetyl cysteine or melatonin. This effect of antioxidant was also true in HFD-fed CKO mice, suppressing fatty liver development and improving hepatic mitochondrial function. Expression of ER stress marker proteins and hepatic fat deposition also increased in normal-diet, aged CKO mice compared to WT mice. These findings suggest that H2O2 production may be an important event triggering NAFLD and that catalase may be an attractive therapeutic target for preventing NAFLD.
Collapse
Affiliation(s)
- Su-Kyung Shin
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Hyun-Woo Cho
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Seung-Eun Song
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Jae-Hoon Bae
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Seung-Soon Im
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, South Korea
| | - Inha Hwang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Women's University, Seoul, 03760, South Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Women's University, Seoul, 03760, South Korea
| | - Dae-Kyu Song
- Department of Physiology & Obesity-mediated Disease Research Center, Keimyung University School of Medicine, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, South Korea.
| |
Collapse
|
24
|
Moneim AA, Abdel-Rehe ES, Helmy H, Addaleel W. Antidiabetic Effect of Thymoquinone via Modulation of PPAR-γ, GLUT4, Hyperlipidemia and Antioxidant Status in Diabetic Rats. ACTA ACUST UNITED AC 2018. [DOI: 10.3923/ajbs.2018.203.209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
25
|
Izuchi R, Ishijima T, Okada S, Abe K, Nakai Y. Hepatic fatty acid biosynthesis in KK-A y mice is modulated by administration of persimmon peel extract: A DNA microarray study. Food Sci Nutr 2018; 6:1657-1663. [PMID: 30258609 PMCID: PMC6145309 DOI: 10.1002/fsn3.728] [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: 12/01/2016] [Revised: 11/14/2017] [Accepted: 06/08/2018] [Indexed: 01/12/2023] Open
Abstract
SCOPE Previously, we showed that the intake of a persimmon peel (PP) extract altered hepatic gene expression associated with the insulin signaling pathway and enhanced tyrosine phosphorylation of insulin receptors in nonobese type 2 diabetic Goto-Kakizaki rats. Our objective was to evaluate the effect of fat-soluble PP extract on obese type 2 diabetic KK-Ay mice with insulin resistance. METHODS AND RESULTS KK-Ay mice were fed a diet mixed with 0.1% of the extract for 8 weeks. The total ketone body levels in the plasma of PP extract-fed mice were significantly lower than those in the normal diet-fed mice. Hepatic nonesterified palmitic acid content was higher in the PP extract-fed mice than in normal diet-fed mice. The hepatic gene expression profiles of the treated mice indicated upregulation of fatty acid synthesis and downregulation of inflammation-associated genes, predicting SREBP-1c and PPARγ activation. CONCLUSION These results suggest that the PP extract enhances hepatic fatty acid synthesis via SREBP-1c and PPARγ, as well as anti-inflammatory activity in KK-Ay mice.
Collapse
Affiliation(s)
| | - Tomoko Ishijima
- Graduate School of Agricultural and Life SciencesThe University of TokyoBunkyo‐kuJapan
| | - Shinji Okada
- Graduate School of Agricultural and Life SciencesThe University of TokyoBunkyo‐kuJapan
| | - Keiko Abe
- Graduate School of Agricultural and Life SciencesThe University of TokyoBunkyo‐kuJapan
- Project on Health and Anti‐agingKanagawa Academy of Science and Technology Life Science & Environment Research CenterKawasakiJapan
| | - Yuji Nakai
- Graduate School of Agricultural and Life SciencesThe University of TokyoBunkyo‐kuJapan
| |
Collapse
|
26
|
Oppermann H, Alvanos A, Seidel C, Meixensberger J, Gaunitz F. Carnosine influences transcription via epigenetic regulation as demonstrated by enhanced histone acetylation of the pyruvate dehydrogenase kinase 4 promoter in glioblastoma cells. Amino Acids 2018; 51:61-71. [DOI: 10.1007/s00726-018-2619-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/12/2018] [Indexed: 01/11/2023]
|
27
|
Park JE, Lee EJ, Kim JK, Song Y, Choi JH, Kang MJ. Flightless-I Controls Fat Storage in Drosophila. Mol Cells 2018; 41:603-611. [PMID: 29890821 PMCID: PMC6030243 DOI: 10.14348/molcells.2018.0120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/04/2018] [Accepted: 05/21/2018] [Indexed: 01/24/2023] Open
Abstract
Triglyceride homeostasis is a key process of normal development and is essential for the maintenance of energy metabolism. Dysregulation of this process leads to metabolic disorders such as obesity and hyperlipidemia. Here, we report a novel function of the Drosophila flightless-I (fliI) gene in lipid metabolism. Drosophila fliI mutants were resistant to starvation and showed increased levels of triglycerides in the fat body and intestine, whereas fliI overexpression decreased triglyceride levels. These flies suffered from metabolic stress indicated by increased levels of trehalose in hemolymph and enhanced phosphorylation of eukaryotic initiation factor 2 alpha (eIF2α). Moreover, upregulation of triglycerides via a knockdown of fliI was reversed by a knockdown of desat1 in the fat body of flies. These results indicate that fliI suppresses the expression of desat1, thereby inhibiting the development of obesity; fliI may, thus, serve as a novel therapeutic target in obesity and metabolic diseases.
Collapse
Affiliation(s)
- Jung-Eun Park
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505,
Korea
| | - Eun Ji Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505,
Korea
| | - Jung Kwan Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919,
Korea
| | - Youngsup Song
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505,
Korea
| | - Jang Hyun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919,
Korea
| | - Min-Ji Kang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505,
Korea
| |
Collapse
|
28
|
Kraakman MJ, Liu Q, Postigo-Fernandez J, Ji R, Kon N, Larrea D, Namwanje M, Fan L, Chan M, Area-Gomez E, Fu W, Creusot RJ, Qiang L. PPARγ deacetylation dissociates thiazolidinedione's metabolic benefits from its adverse effects. J Clin Invest 2018; 128:2600-2612. [PMID: 29589839 DOI: 10.1172/jci98709] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/22/2018] [Indexed: 01/10/2023] Open
Abstract
Thiazolidinediones (TZDs) are PPARγ agonists with potent insulin-sensitizing effects. However, their use has been curtailed by substantial adverse effects on weight, bone, heart, and hemodynamic balance. TZDs induce the deacetylation of PPARγ on K268 and K293 to cause the browning of white adipocytes. Here, we show that targeted PPARγ mutations resulting in constitutive deacetylation (K268R/K293R, 2KR) increased energy expenditure and protected from visceral adiposity and diet-induced obesity by augmenting brown remodeling of white adipose tissues. Strikingly, when 2KR mice were treated with rosiglitazone, they maintained the insulin-sensitizing, glucose-lowering response to TZDs, while displaying little, if any, adverse effects on fat deposition, bone density, fluid retention, and cardiac hypertrophy. Thus, deacetylation appears to fulfill the goal of dissociating the metabolic benefits of PPARγ activation from its adverse effects. Strategies to leverage PPARγ deacetylation may lead to the design of safer, more effective agonists of this nuclear receptor in the treatment of metabolic diseases.
Collapse
Affiliation(s)
| | - Qiongming Liu
- Naomi Berrie Diabetes Center.,Department of Pathology and Cell Biology
| | - Jorge Postigo-Fernandez
- Naomi Berrie Diabetes Center.,Department of Medicine, Columbia Center for Translational Immunology
| | - Ruiping Ji
- Center for Advanced Cardiac Care, Department of Medicine, Division of Cardiology
| | - Ning Kon
- Institute for Cancer Genetics and Department of Pathology, and
| | - Delfina Larrea
- Department of Neurology, College of Physicians and Surgeons, Columbia University New York, New York, USA
| | - Maria Namwanje
- Naomi Berrie Diabetes Center.,Department of Pathology and Cell Biology
| | - Lihong Fan
- Naomi Berrie Diabetes Center.,Department of Pathology and Cell Biology.,Department of Cardiology, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an City, Shaanxi Province, China
| | - Michelle Chan
- Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Estela Area-Gomez
- Department of Neurology, College of Physicians and Surgeons, Columbia University New York, New York, USA
| | - Wenxian Fu
- Department of Pediatrics, UCSD, La Jolla, California, USA
| | - Remi J Creusot
- Naomi Berrie Diabetes Center.,Department of Medicine, Columbia Center for Translational Immunology
| | - Li Qiang
- Department of Pathology and Cell Biology
| |
Collapse
|
29
|
BMPs as new insulin sensitizers: enhanced glucose uptake in mature 3T3-L1 adipocytes via PPARγ and GLUT4 upregulation. Sci Rep 2017; 7:17192. [PMID: 29222456 PMCID: PMC5722815 DOI: 10.1038/s41598-017-17595-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 11/29/2017] [Indexed: 02/08/2023] Open
Abstract
Insulin-resistance is the main cause of type 2 diabetes. Here we describe the identification and characterization of BMP2 and BMP6 as new insulin-sensitizing growth factors in mature adipocytes. We show that BMP2 and BMP6 lead to enhanced insulin-mediated glucose uptake in both insulin-sensitive and -insensitive adipocytes. We exclude a direct effect of BMP2 or BMP6 on translocation of GLUT4 to the plasma membrane and demonstrate that these BMPs increase GLUT4 protein levels equipotent to Rosiglitazone. BMPs induce expression of PPARγ as the crucial mediator for the insulin-sensitizing effect. A comprehensive RNA-Seq analysis in mature adipocytes revealed regulation of both BMP/Smad and PPARγ target genes. The effects of BMP2 and BMP6 are not completely redundant and include regulation of genes involved in glucose and fatty acid metabolism and adipokine expression. Collectively, these findings suggest the BMP2 and BMP6 pathway(s) as promising new drug targets to treat insulin resistance.
Collapse
|
30
|
Chiang MC, Cheng YC, Nicol CJ, Lin CH. The neuroprotective role of rosiglitazone in advanced glycation end product treated human neural stem cells is PPARgamma-dependent. Int J Biochem Cell Biol 2017; 92:121-133. [DOI: 10.1016/j.biocel.2017.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 09/08/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022]
|
31
|
Hosseini A, Salman M, Zhou Z, Drackley JK, Trevisi E, Loor JJ. Level of dietary energy and 2,4-thiazolidinedione alter molecular and systemic biomarkers of inflammation and liver function in Holstein cows. J Anim Sci Biotechnol 2017; 8:64. [PMID: 28781774 PMCID: PMC5537929 DOI: 10.1186/s40104-017-0196-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 07/04/2017] [Indexed: 02/24/2023] Open
Abstract
Background The objective of the study was to evaluate the effect of overfeeding a moderate energy diet and a 2,4-thiazolidinedione (TZD) injection on blood and hepatic tissue biomarkers of lipid metabolism, oxidative stress, and inflammation as it relates to insulin sensitivity. Results Fourteen dry non-pregnant cows were fed a control (CON) diet to meet 100% of NRC requirements for 3 wk, after which half of the cows were assigned to a moderate-energy diet (OVE) and half of the cows continued on CON for 6 wk. All cows received an intravenous injection of 4 mg TZD/kg of body weight (BW) daily from 2 wk after initiation of dietary treatments and for 2 additional week. Compared with CON cows and before TZD treatment, the OVE cows had lower concentration of total protein, urea and albumin over time. The concentration of cholesterol and tocopherol was greater after 2 wk of TZD regardless of diet. Before and after TZD, the OVE cows had greater concentrations of AST/GOT, while concentrations of paraoxonase, total protein, globulin, myeloperoxidase, and haptoglobin were lower compared with CON cows. Regardless of diet, TZD administration increased the concentration of ceruloplasmin, ROMt, cholesterol, tocopherol, total protein, globulin, myeloperoxidase and beta-carotene. In contrast, the concentration of haptoglobin decreased at the end of TZD injection regardless of diet. Prior to TZD injection, the mRNA expression of PC, ANGPTL4, FGF21, INSR, ACOX1, and PPARD in liver of OVE cows was lower compared with CON cows. In contrast, the expression of HMGCS2 was greater in OVE compared with CON cows. After 1 wk of TZD administration the expression of IRS1 decreased regardless of diet; whereas, expression of INSR increased after 2 wk of TZD injection. Cows fed OVE had lower overall expression of TNF, INSR, PC, ACOX1, FGF21, and PPARD but greater HMGCS2 expression. These differences were most evident before and after 1 wk of TZD injection, and by 2 wk of TZD differences in expression for most genes disappeared. Conclusions Based on molecular and blood data, administration of TZD enhanced some aspects of insulin sensitivity while causing contradictory results in terms of inflammation and oxidative stress. The bovine liver is TZD-responsive and level of dietary energy can modify the effects of TZD. Because insulin sensitizers have been proposed as useful tools to manage dairy cows during the transition period, further studies are required to investigate the potential hepatotoxicity effect of TZD (or similar compounds) in dairy cattle. Electronic supplementary material The online version of this article (doi:10.1186/s40104-017-0196-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Afshin Hosseini
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA
| | - Mustafa Salman
- Department of Animal Nutrition and Nutritional Diseases, University of Ondokuz Mayıs, 55139 Samsun, Turkey
| | - Zheng Zhou
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA
| | - James K Drackley
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA
| | - Erminio Trevisi
- Istituto di Zootecnica, Facoltà di Scienze Agrarie Alimentari ed Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Juan J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, 1207 West Gregory Drive, Urbana, IL 61801 USA
| |
Collapse
|
32
|
RORα controls hepatic lipid homeostasis via negative regulation of PPARγ transcriptional network. Nat Commun 2017; 8:162. [PMID: 28757615 PMCID: PMC5534431 DOI: 10.1038/s41467-017-00215-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 06/09/2017] [Indexed: 01/15/2023] Open
Abstract
The retinoic acid receptor-related orphan receptor-α (RORα) is an important regulator of various biological processes, including cerebellum development, circadian rhythm and cancer. Here, we show that hepatic RORα controls lipid homeostasis by negatively regulating transcriptional activity of peroxisome proliferators-activated receptor-γ (PPARγ) that mediates hepatic lipid metabolism. Liver-specific Rorα-deficient mice develop hepatic steatosis, obesity and insulin resistance when challenged with a high-fat diet (HFD). Global transcriptome analysis reveals that liver-specific deletion of Rorα leads to the dysregulation of PPARγ signaling and increases hepatic glucose and lipid metabolism. RORα specifically binds and recruits histone deacetylase 3 (HDAC3) to PPARγ target promoters for the transcriptional repression of PPARγ. PPARγ antagonism restores metabolic homeostasis in HFD-fed liver-specific Rorα deficient mice. Our data indicate that RORα has a pivotal role in the regulation of hepatic lipid homeostasis. Therapeutic strategies designed to modulate RORα activity may be beneficial for the treatment of metabolic disorders. Hepatic steatosis development may result from dysregulation of lipid metabolism, which is finely tuned by several transcription factors including the PPAR family. Here Kim et al. show that the nuclear receptor RORα inhibits PPARγ-mediated transcriptional activity by interacting with HDAC3 and competing for the promoters of lipogenic genes.
Collapse
|
33
|
Man K, Kutyavin VI, Chawla A. Tissue Immunometabolism: Development, Physiology, and Pathobiology. Cell Metab 2017; 25:11-26. [PMID: 27693378 PMCID: PMC5226870 DOI: 10.1016/j.cmet.2016.08.016] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
Abstract
Evolution of metazoans resulted in the specialization of cellular and tissue function. This was accomplished by division of labor, which allowed tissue parenchymal cells to prioritize their core functions while ancillary functions were delegated to tissue accessory cells, such as immune, stromal, and endothelial cells. In metabolic organs, the accessory cells communicate with their clients, the tissue parenchymal cells, to optimize cellular processes, allowing organisms to adapt to changes in their environment. Here, we discuss tissue immunometabolism from this vantage point and use examples from adipose tissues (white, beige, and brown) and liver to outline the general principles by which accessory cells support metabolic homeostasis in parenchymal cells. A corollary of this model is that disruption of communication between client and accessory cells might predispose metabolic organs to the development of disease.
Collapse
Affiliation(s)
- Kevin Man
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Vassily I Kutyavin
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Ajay Chawla
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA; Departments of Physiology and Medicine, University of California, San Francisco, CA 94143-0795, USA.
| |
Collapse
|
34
|
The effects of PPARγ on the regulation of the TOMM40-APOE-C1 genes cluster. Biochim Biophys Acta Mol Basis Dis 2017; 1863:810-816. [PMID: 28065845 DOI: 10.1016/j.bbadis.2017.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/12/2016] [Accepted: 01/04/2017] [Indexed: 11/24/2022]
Abstract
Chromosome 19q13.32 is a gene rich region, and has been implicated in multiple human phenotypes in adulthood including lipids traits, Alzheimer's disease, and longevity. Peroxisome Proliferator Activated Receptor Gamma (PPARγ) is a ligand-activated nuclear transcription factor that plays a role in human complex traits that are also genetically associated with the chromosome 19q13.32 region. Here, we study the effects of PPARγ on the regional expression regulation of the genes clustered within chromosome 19q13.32, specifically TOMM40, APOE, and APOC1, applying two complementary approaches. Using the short hairpin RNA (shRNA) method in the HepG2 cell-line we knocked down PPARγ expression and measured the effect on mRNA expression. We discovered PPARγ knock down increased the levels of TOMM40-, APOE-, and APOC1-mRNAs, with the highest increase in expression observed for APOE-mRNA. To complement the PPARγ knockdown findings we also examined the effects of low doses of PPARγ agonists (nM range) on mRNA expression of these genes. Low (nM) concentrations of pioglitazone (Pio) decreased transcription of TOMM40, APOE, and APOC1 genes, with the lowest mRNA levels for each gene observed at 1.5nM. Similar to the effect of PPARγ knockdown, the strongest response to pioglitazone was also observed for APOE-mRNA, and rosiglitazone (Rosi), another PPARγ agonist, produced results that were consistent with these. In conclusion, our results further established a role for PPARγ in regional transcriptional regulation of chr19q13.32, underpinning the association between PPARγ, the chr19q13.32 genes cluster, and human complex traits and disease.
Collapse
|
35
|
Pichiah PBT, Cho SH, Han SK, Cha YS. Fermented Barley Supplementation Modulates the Expression of Hypothalamic Genes and Reduces Energy Intake and Weight Gain in Rats. J Med Food 2016; 19:418-26. [PMID: 27074621 DOI: 10.1089/jmf.2015.3600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Dietary fiber and proteins are individually known to decrease feeding, but could result greater weight management benefit when both are combined. We hypothesized that supplementing the diet with fermented barley, being rich in both dietary fiber and proteins, could lower energy intake by modulating the mRNA expression level of hypothalamic genes associated with the regulation of feeding behavior and satiety; thereby decreasing body weight gain. To test our hypothesis, four groups of Sprague Dawley rats were arranged in a 2 × 2 factorial design (n = 6), low-fat diet with either guar gum (LFD-G) or fermented barley (LFD-FB) and high-fat diet with either guar gum (HFD-G) or fermented barley (HFD-FB). Using oral gavage, fermented barley was given at a dosage of 1500 mg/kg body weight and guar gum was supplemented in an equivalent quantity to that of the fiber in the fermented barley. After 19 weeks, the fermented barley-supplemented groups showed a significant reduction in energy intake, triglyceride, body weight gain, and serum leptin, compared to the guar gum-supplemented groups in both the low- and high-fat diet groups. Likewise, the anorexigenic gene proopiomelanocortin (POMC) and cocaine and amphetamine-regulated transcript (CART) mRNA level were significantly higher in the fermented barley-supplemented groups compared to the guar gum-supplemented groups in rats fed on both high- and low-fat diets. In conclusion, fermented barley supplementation upregulated hypothalamic POMC/CART, decreased energy intake in both low- and high-fat diet groups, and prevented excessive weight gain in rats.
Collapse
Affiliation(s)
- P B Tirupathi Pichiah
- 1 Department of Food Science and Human Nutrition, Chonbuk National University , Jeonju, Korea
| | - Suk-Ho Cho
- 2 Mealtobalance, Co., Ltd. , Jeongeup, Jeonbuk, Korea
| | - Seong-Kyu Han
- 3 Department of Oral Physiology School of Dentistry and Institute of Oral Bioscience, Chonbuk National University , Jeonju, Korea
| | - Youn-Soo Cha
- 1 Department of Food Science and Human Nutrition, Chonbuk National University , Jeonju, Korea.,4 AgroBiofood R&D Institute of Chonbuk National University , Jeonju, Korea
| |
Collapse
|
36
|
Martins FO, Delgado TC, Viegas J, Gaspar JM, Scott DK, O'Doherty RM, Macedo MP, Jones JG. Mechanisms by which the thiazolidinedione troglitazone protects against sucrose-induced hepatic fat accumulation and hyperinsulinaemia. Br J Pharmacol 2016; 173:267-78. [PMID: 26447327 DOI: 10.1111/bph.13362] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 08/13/2015] [Accepted: 09/29/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Thiazolidinediones (TZD) are known to ameliorate fatty liver in type 2 diabetes. To date, the underlying mechanisms of their hepatic actions remain unclear. EXPERIMENTAL APPROACH Hepatic triglyceride content and export rates were assessed in 2 week high-sucrose-fed Wistar rats treated with troglitazone and compared with untreated high-sucrose rodent controls. Fractional de novo lipogenesis (DNL) contributions to hepatic triglyceride were quantified by analysis of triglyceride enrichment from deuterated water. Hepatic insulin clearance and NO status during a meal tolerance test were also evaluated. KEY RESULTS TZD significantly reduced hepatic triglyceride (P < 0.01) by 48%, decreased DNL contribution to hepatic triglyceride (P < 0.01) and increased postprandial non-esterified fatty acids clearance rates (P < 0.01) in comparison with the high-sucrose rodent control group. During a meal tolerance test, plasma insulin AUC was significantly lower (P < 0.01), while blood glucose and plasma C-peptide levels were not different. Insulin clearance was increased (P < 0.001) by 24% and was associated with a 22% augmentation of hepatic insulin-degrading enzyme activity (P < 0.05). Finally, hepatic NO was decreased by 24% (P < 0.05). CONCLUSIONS Overall, TZD show direct actions on liver by reducing hepatic DNL and increasing hepatic insulin clearance. The alterations in hepatic insulin clearance were associated with changes in insulin-degrading enzyme activity, with possible modulation of NO levels.
Collapse
Affiliation(s)
- Fátima O Martins
- Metabolic Control Group, Center for Neurosciences and Cell Biology of Coimbra, Cantanhede, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Miyazaki Y, De Filippis E, Bajaj M, Wajcberg E, Glass L, Triplitt C, Cersosimo E, Mandarino LJ, Defronzo RA. Predictors of improved glycaemic control with rosiglitazone therapy in type 2 diabetic patients: a practical approach for the primary care physician. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/14746514050050010601] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Objective — To determine predictors of improved glycaemic control in patients with type 2 diabetes mellitus during rosiglitazone therapy using basic clinical parameters that are readily available in daily clinical practice. Research design and methods — Thirty-seven type 2 diabetic patients (men/women = 18/19; age = 54±2years; diabetes duration = 6±1 years; diet-/sulphonylurea-treated = 24/13) received a 75 g oral glucose tolerance test (OGTT) and determination of body fat before and after rosiglitazone (8 mg/day) for 12 weeks. Results — After rosiglitazone therapy, there were decreases in HbA1C (8.6±0.2 to 7.2±0.2%, p<0.0001), fasting plasma glucose (FPG) (10.6±0.3 to 8.0±0.3 mmol/L [191±6 to 145±6 mg/dL], p<0.0001), fasting plasma insulin (FPI) (108±6 to 84±6 pmol/L [18±1 to 14±µU/ml], p<0.05), fasting free fatty acids (FFA) (760±39 to 611±33 µEq/l, p<0.05), mean plasma glucose (PG) — OGTT (16.2±0.39 to 12.7±0.33 mmol/L [291±7 to 230±6 mg/dL], p<0.001), and mean FFA-OGTT (604±27 to 445±23 µEq/l, p<0.01) despite increases in body weight (85±2 to 88±2 kg, p<0.01) and % fat mass (37.9±2.0 to 39.5±1.9%, p<0.01). The insulinogenic index (IGI) during 0—120 minutes OGTT (IGI0-120) increased following rosiglitazone (0.19±0.03 to 0.30±0.05, p<0.01). Two different insulin sensitivity indices, calculated from PG and plasma insulin (PI) during OGTT, increased significantly: composite index of whole body insulin sensitivity (ISIcomposite): 2.3±0.3 to 3.4±0.4, p<0.05; oral glucose insulin sensitivity (OGIS): 248±5 to 294±6 ml/m2.min, p<0.001. Using clinical and laboratory variables obtained in daily clinical practice (age, gender, diabetes duration, sulphonylurea treatment, body mass index (BMI), % fat mass, fasting plasma insulin/C-peptide/FFA/lipids, IGI0-30, IGI0-120, and ISIcomposite or OGIS), stepwise regression analysis demonstrated that % fat mass (standard coefficient (S.C.) = —0.49, p=0.001) and OGIS (S.C. = 0.31, p=0.02) or ISIcomp (S.C. = 0.31, p=0.03) are significant predictors of the decrease in HbA1C after rosiglitazone (adjusted R2 =0.33, p=0.0004). Conclusions — Rosiglitazone improves insulin resistance and glycaemic control in type 2 diabetes. Obesity (more body fat mass) and reduced insulin sensitivity prior to treatment are the best predictors of glycaemic response to thiazolidinedione therapy in type 2 diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ralph A Defronzo
- University of Texas Health Science Center, Diabetes Division, Room 3.380S, 7703 Floyd Curl Drive, San Antonio, Texas, 78229-3900, USA,
| |
Collapse
|
38
|
Eny KM, El-Sohemy A, Cornelis MC, Sung YK, Bae SC. Catalase and PPARg2 genotype and risk of systemic lupus erythematosus in Koreans. Lupus 2016; 14:351-5. [PMID: 15934434 DOI: 10.1191/0961203305lu2091oa] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Catalase (CAT) and peroxisome proliferator activated receptor-g2 (PPARg2) are important regulators of oxidative stress and inflammation, which may contribute to the development of systemic lupus erythematosus (SLE). The objective of this study was to investigate the effects of genetic polymorphisms of CAT and PPARg2 on risk and severity of SLE in a Korean population. DNA was isolated from blood samples collected from 345 patients with SLE and 400 controls. Genotyping for the 2262C!T polymorphism of CAT and the Pro12Ala polymorphism of PPARg2 were performed by PCR-RFLP analysis. The severity of SLE was assessed using the Systemic Lupus International Collaborating Clinics/American College of Rheumatology (SLICC/ACR) damage index (SDI). No association was observed between genotypes for any of the clinical manifestations of SLE. CAT and PPARg2 genotypes were not associated with either risk or severity of SLE. For subjects who were carriers of the high activity Tallele for CATand have the Pro/Pro genotype for PPARg2, the odds ratio (95% confidence interval) for risk of SLE was 0.45 (0.23-1.08). Our results suggest that genetic polymorphisms of CAT and PPARg2 do not play a significant role in the development of SLE in a Korean population. A possible protective effect of a combined genotype warrants further investigation.
Collapse
Affiliation(s)
- K M Eny
- Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| | | | | | | | | |
Collapse
|
39
|
Walton RG, Zhu X, Tian L, Heywood EB, Liu J, Hill HS, Liu J, Bruemmer D, Yang Q, Fu Y, Garvey WT. AP2-NR4A3 transgenic mice display reduced serum epinephrine because of increased catecholamine catabolism in adipose tissue. Am J Physiol Endocrinol Metab 2016; 311:E69-81. [PMID: 27166283 PMCID: PMC4967153 DOI: 10.1152/ajpendo.00330.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 05/03/2016] [Indexed: 01/25/2023]
Abstract
The NR4A orphan nuclear receptors function as early response genes to numerous stimuli. Our laboratory has previously demonstrated that overexpression of NR4A3 (NOR-1, MINOR) in 3T3-L1 adipocytes enhances insulin-stimulated glucose uptake. To assess the in vivo effect of NR4A3 on adipocytes, we generated transgenic mice with NR4A3 overexpression driven by the adipocyte fatty acid-binding protein (AP2) promoter (AP2-NR4A3 mice). We hypothesized that AP2-NR4A3 mice would display enhanced glucose tolerance and insulin sensitivity. However, AP2-NR4A3 mice exhibit metabolic impairment, including increased fasting glucose and insulin, impaired glucose tolerance, insulin resistance, decreased serum free fatty acids, and increased low-density lipoprotein-cholesterol. AP2-NR4A3 mice also display a significant reduction in serum epinephrine due to increased expression of catecholamine-catabolizing enzymes in adipose tissue, including monoamine oxidase-A. Furthermore, enhanced expression of monoamine oxidase-A is due to direct transcriptional activation by NR4A3. Finally, AP2-NR4A3 mice display cardiac and behavioral alterations consistent with chronically low circulating epinephrine levels. In conclusion, overexpression of NR4A3 in adipocytes produces a complex phenotype characterized by impaired glucose metabolism and low serum catecholamines due to enhanced degradation by adipose tissue.
Collapse
Affiliation(s)
- R Grace Walton
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama;
| | - Xiaolin Zhu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ling Tian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elizabeth B Heywood
- Saha Cardiovascular Research Center and Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Jian Liu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Helliner S Hill
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jiarong Liu
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Dennis Bruemmer
- Saha Cardiovascular Research Center and Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Qinglin Yang
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yuchang Fu
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - W Timothy Garvey
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama; Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
40
|
Liu L, Lin Y, Liu L, Wang L, Bian Y, Gao X, Li Q. Regulation of peroxisome proliferator-activated receptor gamma on milk fat synthesis in dairy cow mammary epithelial cells. In Vitro Cell Dev Biol Anim 2016; 52:1044-1059. [DOI: 10.1007/s11626-016-0059-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 05/18/2016] [Indexed: 12/26/2022]
|
41
|
Pershadsingh HA. Dual Peroxisome Proliferator-Activated Receptor-alpha/gamma Agonists : In the Treatment of Type 2 Diabetes Mellitus and the Metabolic Syndrome. ACTA ACUST UNITED AC 2016; 5:89-99. [PMID: 16542049 DOI: 10.2165/00024677-200605020-00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The metabolic syndrome consists of a combination of cardiovascular risk factors that include hyperglycemia with or without type 2 diabetes mellitus, visceral obesity, elevated blood pressure, and atherogenic dyslipidemia. These interrelated disorders and their associated lipotoxicity, oxidative stress, and inflammatory state predispose to a constellation of cardiovascular conditions leading to high risk of heart attack, stroke, renal failure, blindness, and lower extremity amputation. Visceral obesity, a prime risk factor for type 2 diabetes and a major component of the metabolic syndrome, potentiates atherogenesis, atherosclerosis, organ lipotoxicity, and oxidative tissue damage.Peroxisome proliferator-activated receptors (PPARs) are relatively recently discovered nuclear transcription factors that are modulated by dietary fatty acids, including the essential polyunsaturated fatty acids, arachidonic acid and its metabolites, and are essential to the control of energy metabolism. Of the three PPAR isoforms (alpha, gamma, and delta), synthetic pharmaceutical ligands that activate PPARalpha (the antidyslipidemic fibric acid derivatives ['fibrates']) and PPARgamma (the antidiabetic thiazolidinediones) have been studied extensively. Recently developed dual PPARalpha/gamma agonists may combine the therapeutic effects of these drugs, creating the expectation of greater efficacy, and perhaps other advantages in the treatment of type 2 diabetes and the metabolic syndrome. However, thiazolidinediones are hampered by adverse effects related to increased weight gain and fluid overload. It remains to be seen whether the dual PPARalpha/gamma agonists currently under development have similar limitations. Nevertheless, existing clinical data imply that the combined effects of thiazolidinediones and fibrates are likely to be emulated by dual PPARalpha/gamma agonists, providing superior efficacy to these classes for the treatment of type 2 diabetes, the metabolic syndrome, and their cardiovascular and other end-organ complications.
Collapse
|
42
|
Zhang W, Sun Q, Zhong W, Sun X, Zhou Z. Hepatic Peroxisome Proliferator-Activated Receptor Gamma Signaling Contributes to Alcohol-Induced Hepatic Steatosis and Inflammation in Mice. Alcohol Clin Exp Res 2016; 40:988-99. [PMID: 27062444 DOI: 10.1111/acer.13049] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/18/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor gamma (PPARγ) signaling has been shown to regulate lipogenesis and lipid accumulation. Previous studies have shown that hepatic PPARγ is up-regulated in steatotic liver of both animal and human. However, the effects of hepatic PPARγ signaling on alcoholic liver disease (ALD) remain elusive. METHODS To determine the role of hepatic PPARγ signaling on ALD, wild-type (WT) and hepatocyte-specific PPARγ knockdown (PPARγ∆Hep) mice were fed a modified Lieber-DeCarli alcohol or isocaloric maltose dextrin control liquid diet for 8 weeks to induce ALD. Blood parameters, hepatic steatosis, and inflammation were measured after 8-week alcohol feeding. RESULTS Alcohol feeding to WT mice resulted in liver damage (alanine aminotransferase [ALT], 94.68 ± 17.05 U/L; aspartate aminotransferase [AST], 55.87 ± 11.29 U/L), which was significantly alleviated by hepatic PPARγ knockdown (ALT, 57.36 ± 14.98 U/L; AST, 38.06 ± 3.35 U/L). Alcohol feeding led to marked lipid accumulation and up-regulation of lipogenic genes including fatty acid transport protein 1 (FATP1), acetyl-CoA carboxylase (ACC), fatty acid synthase (FASN), lipin1 (LIPIN1), diacylglycerol acyltransferase 1 (DGAT1), and diacylglycerol acyltransferase 2 (DGAT2) in the livers of WT mice. Knockdown of hepatic PPARγ significantly alleviated alcohol-induced lipid accumulation and abolished the up-regulation of FASN, DGAT1, and DGAT2. Silencing of PPARγ in FL83B cells significantly decreased ethanol (EtOH)-, linoleic acid-, and EtOH plus linoleic acid-induced lipid accumulation. Knockdown of hepatic PPARγ also significantly reduced alcohol-induced inflammatory chemokine (monocyte chemotactic protein 1 [MCP1], keratinocyte-derived chemokine [KC], interferon gamma-induced protein 10 [IP-10]) and inflammatory infiltration (lymphocyte antigen 6 complex, locus G [Ly6G], and F4/80). CONCLUSIONS The results suggest that hepatic PPARγ signaling contributes to alcohol-induced liver injury by promoting hepatic steatosis and inflammation.
Collapse
Affiliation(s)
- Wenliang Zhang
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Qian Sun
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina.,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Wei Zhong
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Xinguo Sun
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina.,Department of Nutrition, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| |
Collapse
|
43
|
Kido H, Indalao IL, Kim H, Kimoto T, Sakai S, Takahashi E. Energy metabolic disorder is a major risk factor in severe influenza virus infection: Proposals for new therapeutic options based on animal model experiments. Respir Investig 2016; 54:312-9. [PMID: 27566378 DOI: 10.1016/j.resinv.2016.02.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 12/14/2022]
Abstract
Severe influenza is characterized by cytokine storm and multiorgan failure. Influenza patients with underlying diseases show a rapid progression in disease severity. The major mechanism that underlies multiorgan failure during the progressive stage of infection, particularly in patients with underlying risk factors, is mitochondrial energy crisis. The relationship between the factors that determine infection severity, such as influenza virus, cytokines, cellular trypsin as a hemagglutinin processing protease for viral multiplication, accumulation of metabolic intermediates and ATP crisis in mitochondria, is termed the "influenza virus-cytokine-trypsin" cycle. This occurs during the initial stages of infection, and is interconnected with the "metabolic disorders-cytokine" cycle in the middle to late phase of infection. Experiments using animal models have highlighted the complex relationship between these two cycles. New treatment options have been proposed that target the ATP crisis and multiorgan failure during the late phase of infection, rather than antiviral treatments with neuraminidase inhibitors that work during the initial phase. These options are (i) restoration of glucose oxidation in mitochondria by diisopropylamine dichloroacetate, which inhibits infection-induced pyruvate dehydrogenase kinase 4 activity, and (ii) restoration of long-chain fatty acid oxidation in mitochondria by l-carnitine and bezafibrate, an agonist of peroxisome proliferation-activated receptors-β/δ, which transcriptionally upregulates carnitine palmitoyltransferase II. The latter is particularly effective in patients with influenza-associated encephalopathy who have thermolabile and short half-life compound variants of carnitine palmitoyltransferase II.
Collapse
Affiliation(s)
- Hiroshi Kido
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Kuramoto-cho 3-18-15, Tokushima 770-8503, Japan.
| | - Irene L Indalao
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Kuramoto-cho 3-18-15, Tokushima 770-8503, Japan.
| | - Hyejin Kim
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Kuramoto-cho 3-18-15, Tokushima 770-8503, Japan.
| | - Takashi Kimoto
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Kuramoto-cho 3-18-15, Tokushima 770-8503, Japan.
| | - Satoko Sakai
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Kuramoto-cho 3-18-15, Tokushima 770-8503, Japan.
| | - Etsuhisa Takahashi
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Kuramoto-cho 3-18-15, Tokushima 770-8503, Japan.
| |
Collapse
|
44
|
Husi H, Human C. Molecular determinants of acute kidney injury. J Inj Violence Res 2016; 7:75-86. [PMID: 26104320 PMCID: PMC4522318 DOI: 10.5249/jivr.v7i2.615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 07/10/2014] [Indexed: 12/18/2022] Open
Abstract
Background: Acute kidney injury (AKI) is a condition that leads to a rapid deterioration of renal function associated with impairment to maintain electrolyte and acid balance, and, if left untreated, ultimately irreversible kidney damage and renal necrosis. There are a number of causes that can trigger AKI, ranging from underlying conditions as well as trauma and surgery. Specifically, the global rise in surgical procedures led to a substantial increase of AKI incidence rates, which in turn impacts on mortality rates, quality of life and economic costs to the healthcare system. However, no effective therapy for AKI exists. Current approaches, such as pharmacological intervention, help in alleviating symptoms in slowing down the progression, but do not prevent or reverse AKI-induced organ damage. Methods: An in-depth understanding of the molecular machinery involved in and modulated by AKI induction and progression is necessary to specifically pharmacologically target key molecules. A major hurdle to devise a successful strategy is the multifactorial and complex nature of the disorder itself, whereby the activation of a number of seemingly independent molecular pathways in the kidney leads to apoptotic and necrotic events. Results: The renin-angiotensin-aldosterone-system (RAAS) axis appears to be a common element, leading to downstream events such as triggers of immune responses via the NFB pathway. Other pathways intricately linked with AKI-induction and progression are the tumor necrosis factor alpha (TNF α) and transforming growth factor beta (TGF β) signaling cascades, as well as a number of other modulators. Surprisingly, it has been shown that the involvement of the glutamatergic axis, believed to be mainly a component of the neurological system, is also a major contributor. Conclusions: Here we address the current understanding of the molecular pathways evoked in AKI, their interplay, and the potential to pharmacologically intervene in the effective prevention and/or progression of AKI.
Collapse
Affiliation(s)
- Holger Husi
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK.
| | | |
Collapse
|
45
|
Abstract
Salacia species plant has been used traditionally as an Ayurvedic medicine for diabetes mellitus. Studies over the past decades have shown its multi-targeted role in diabetics. In the present review article, various mechanisms of action of Salacia on diabetics are discussed in detail. Apart from the well-known action of decreasing postprandial glucose sugar by inhibiting α-glucosidase and α-pancreatic amylase, it also inhibits aldose reductase which otherwise results in microvascular complications. Importantly, its peroxisome proliferator-activated receptor (PPAR)-γ agonist (such as thiazolidinediones, the insulin sensitizers) action increases the uptake of free fatty acid (FFA) and facilitates their storage in subcutaneous fat rather than the visceral fat. This reduces plasma FFA and insulin resistance. Furthermore, it increases the expression of and translocation to the cell surface of glucose transporter 1 and 4 receptors which result in glucose uptake by the liver and skeletal muscle and decreases plasma glucose levels. It also decreases inflammatory cytokines and increases adiponectin expression. Salacia as PPAR-α agonist (such as fibrates) has a role in the management of dyslipidemia. The activation of PPAR-α leads to the increased expression of lipoprotein lipase and apolipoprotein (Apo) A-V and decrease in hepatic Apo-C-III. These actions lower plasma triglycerides in chylomicrons and very low-density lipoprotein particles, thus liberating fatty acids, which are taken up and stored as fat in adipocytes. Salacia has been shown to suppress the overexpression of cardiac PPAR-α (similar to angiotensin-converting enzyme inhibitors/angiotensin receptor blockers) and thereby preventing diabetic cardiomyopathy. It also suppresses the cardiac angiotensin II Type 1 receptors resulting in antihypertrophic and antifibrogenic effect.
Collapse
Affiliation(s)
- Neera Vyas
- Assistant Director (Med.), Central Council for Research in Ayurvedic Sciences, Janakpuri, New Delhi, India
| | - Rakhi Mehra
- Department of Clinical research, Central Ayurveda Research Institute for Cardio Vascular Diseases, Punjabi Bagh, New Delhi, India
| | - Renu Makhija
- Department of Clinical research, Central Ayurveda Research Institute for Cardio Vascular Diseases, Punjabi Bagh, New Delhi, India
| |
Collapse
|
46
|
Abstract
Obesity and type 2 diabetes mellitus (T2DM) epidemics, which have already spread, imply the possibility of both conditions being closely related. Thus, the goal of the present review was to draw a parallel between obesity, adipose tissue (AT) changes, and T2DM development. To this end, a search was conducted in PubMed, MEDLINE and SciELO databases, using the following key words and their combinations: obesity; diabetes; insulin resistance; diet; weight loss; adipocin; inflammation markers; and interleukins. Based on a literature review, AT dysfunction observed in obesity is characterised by adipocyte hypertrophy, macrophage infiltration, impaired insulin signalling and insulin resistance. In addition, there is release of inflammatory adipokines and an excessive amount of NEFA promoting ectopic fat deposition and lipotoxicity in muscle, liver and pancreas. Recent evidence supports the hypothesis that the conception of AT as a passive energy storage organ should be replaced by a dynamic endocrine organ, which regulates metabolism through a complex adipocyte communication with the surrounding microenvironment. The present review demonstrates how glucose homeostasis is changed by AT dysfunction. A better understanding of this relationship enables performing nutritional intervention strategies with the goal of preventing T2DM.
Collapse
|
47
|
Lei L, Liu Q, Liu S, Huan Y, Sun S, Chen Z, Li L, Feng Z, Li Y, Shen Z. Antidiabetic potential of a novel dual-target activator of glucokinase and peroxisome proliferator activated receptor-γ. Metabolism 2015; 64:1250-61. [PMID: 26189598 DOI: 10.1016/j.metabol.2015.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 06/17/2015] [Accepted: 06/18/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND PURPOSE Glucokinase (GK) balances blood glucose levels via regulation of glucose metabolism and insulin secretion. Peroxisome proliferator activated receptor-γ (PPARγ) regulates gene expression in glucose and lipid metabolism. In this study, we investigated the therapeutic effect of a novel compound, SHP289-03, which activates both GK and PPARγ. METHODS Glucose metabolism was tested in primary hepatocytes of normal ICR mice, and insulin secretion was measured in NIT-1 insulinoma cells as well as in primary islets of normal ICR mice. The in vivo pharmacodynamics of SHP289-03 was assessed using the spontaneous type 2 diabetic mouse model, KKA(y). KEY RESULTS In hepatocytes, SHP289-03 promoted glucose consumption. In NIT-1 cells, it increased the concentration of intracellular ATP and calcium, and subsequently enhanced glucose-stimulated insulin secretion in both NIT-1 cells and primary islets. Moreover, SHP289-03 decreased the blood glucose level, improved glucose tolerance and reduced blood lipid levels in KKA(y) mice. It restored islet morphology and increased the beta cell/alpha cell mass ratio, in addition to up-regulating GK gene expression in the liver of KKA(y) mice. DISCUSSION AND CONCLUSIONS SHP289-03 has significant therapeutic potential for the treatment of diabetes mellitus.
Collapse
Affiliation(s)
- Lei Lei
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Quan Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Shuainan Liu
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Yi Huan
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Sujuan Sun
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Zhiyu Chen
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Linyi Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Zhiqiang Feng
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Yan Li
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China
| | - Zhufang Shen
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P. R. China.
| |
Collapse
|
48
|
Zhou L, Panasiuk A, Downton M, Zhao D, Yang B, Jia Z, Yang T. Systemic PPARγ deletion causes severe disturbance in fluid homeostasis in mice. Physiol Genomics 2015; 47:541-7. [PMID: 26330489 DOI: 10.1152/physiolgenomics.00066.2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/27/2015] [Indexed: 01/29/2023] Open
Abstract
The pharmacological action of peroxisome proliferator-activated receptor (PPAR)γ in promoting sodium and water retention is well documented as highlighted by the major side-effect of body weight gain and edema associated with thiazolidinedione use. However, a possible physiological role of PPARγ in regulation of fluid metabolism has not been reported by previous studies. Here we analyzed fluid metabolism in inducible whole-body PPARγ knockout mice. The null mice developed severe polydipsia and polyuria, reduced urine osmolality, and modest hyperphagia. The phenomenon persisted during 3 days of pair feeding and pair drinking, accompanied by progressive weight loss. After 24 h water deprivation, the null mice had a lower urine osmolality, a higher urine volume, a greater weight loss, and a greater rise in hematocrit than the floxed control. Urinary vasopressin (AVP) excretion was not different between the genotypes under basal condition or after WD. The response of urine osmolality to acute and chronic 1-desamino-8-D-arginine vasopressin treatment was attenuated in the null mice, but the total abundance or phosphorylation of aquaporin 2 (AQP2) in the kidney or AVP-induced cAMP production in inner medullary collecting duct suspensions was unaffected. Overall, PPARγ participates in physiological control of fluid homeostasis through an unknown mechanism involving cAMP/AQP2-independent enhancement of AVP response.
Collapse
Affiliation(s)
- Li Zhou
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Alexandra Panasiuk
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Maicy Downton
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Daqiang Zhao
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Baoxue Yang
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhanjun Jia
- Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| | - Tianxin Yang
- Institute of Hypertension, Sun Yat-sen University School of Medicine, Guangzhou, China; Internal Medicine, University of Utah and Veterans Affairs Medical Center, Salt Lake City, Utah; and
| |
Collapse
|
49
|
Xue B, Nie J, Wang X, DuBois DC, Jusko WJ, Almon RR. Effects of High Fat Feeding on Adipose Tissue Gene Expression in Diabetic Goto-Kakizaki Rats. GENE REGULATION AND SYSTEMS BIOLOGY 2015; 9:15-26. [PMID: 26309393 PMCID: PMC4533846 DOI: 10.4137/grsb.s25172] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 05/24/2015] [Accepted: 06/17/2015] [Indexed: 12/15/2022]
Abstract
Development and progression of type 2 diabetes is a complex interaction between genetics and environmental influences. High dietary fat is one environmental factor that is conducive to the development of insulin-resistant diabetes. In the present report, we compare the responses of lean poly-genic, diabetic Goto-Kakizaki (GK) rats to those of control Wistar-Kyoto (WKY) rats fed a high fat diet from weaning to 20 weeks of age. This comparison included a wide array of physiological measurements along with gene expression profiling of abdominal adipose tissue using Affymetrix gene array chips. Animals of both strains fed a high fat diet or a normal diet were sacrificed at 4, 8, 12, 16, and 20 weeks for this comparison. The microarray analysis revealed that the two strains developed different adaptations to increased dietary fat. WKY rats decrease fatty acid synthesis and lipogenic processes whereas GK rats increase lipid elimination. However, on both diets the major differences between the two strains remained essentially the same. Specifically relative to the WKY strain, the GK strain showed lipoatrophy, chronic inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Bai Xue
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jing Nie
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Xi Wang
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Debra C DuBois
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, USA
| | - Richard R Almon
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, USA. ; New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY, USA
| |
Collapse
|
50
|
N-3 Polyunsaturated Fatty Acids and Inflammation in Obesity: Local Effect and Systemic Benefit. BIOMED RESEARCH INTERNATIONAL 2015; 2015:581469. [PMID: 26339623 PMCID: PMC4538411 DOI: 10.1155/2015/581469] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/11/2015] [Indexed: 12/19/2022]
Abstract
Overwhelming consensus emerges among countless evidences that obesity is characterized by a chronic low-grade inflammation in the adipose tissue (AT), which subsequently develops into a systemic inflammatory state contributing to obesity-associated diseases. N-3 Polyunsaturated fatty acids (n-3 PUFA), known as important modulators participating in inflammatory process, turn out to be an effective mitigating strategy dealing with local and systemic inflammation observed in obesity. Some of the effects of n-3 PUFA are brought about by regulation of gene expression through interacting with nuclear receptors and transcription factors; other effects are elicited by modulation of the amount and type of mediator derived from PUFAs. The metabolic effects of n-3 PUFA mainly result from their interactions with several organ systems, not limited to AT. Notably, the attenuation of inflammation in hard-hit AT, in turn, contributes to reducing circulating concentrations of proinflammatory cytokines and detrimental metabolic derivatives, which is beneficial for the function of other involved organs. The present review highlights a bridging mechanism between n-3 PUFA-mediated inflammation relief in AT and systemic benefits.
Collapse
|