1
|
Ahn C, Zhang T, Yang G, Rode T, Varshney P, Ghayur SJ, Chugh OK, Jiang H, Horowitz JF. Years of endurance exercise training remodel abdominal subcutaneous adipose tissue in adults with overweight or obesity. Nat Metab 2024; 6:1819-1836. [PMID: 39256590 DOI: 10.1038/s42255-024-01103-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024]
Abstract
Abnormalities in the structure and metabolic function of abdominal subcutaneous adipose tissue (aSAT) underlie many obesity-related health complications. Endurance exercise improves cardiometabolic health in adults with overweight or obesity, but the effects of endurance training on aSAT are unclear. We included male and female participants who were regular exercisers with overweight or obesity who exercised for >2 years, and cross-sectionally compared them with well-matched non-exercisers with overweight or obesity. Here we show aSAT from exercisers has a higher capillary density, lower Col6a abundance and fewer macrophages compared with non-exercisers. This is accompanied by a greater abundance of angiogenic, ribosomal, mitochondrial and lipogenic proteins. The abundance of phosphoproteins involved in protein translation, lipogenesis and direct regulation of transcripts is also greater in aSAT collected from exercisers. Exploratory ex vivo experiments demonstrate greater angiogenic capacity and higher lipid-storage capacity in samples cultured from aSAT collected from exercisers versus non-exercisers. Regular exercise may play a role in remodelling aSAT structure and proteomic profile in ways that may contribute to preserved cardiometabolic health.
Collapse
Affiliation(s)
- Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Tao Zhang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Gayoung Yang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Thomas Rode
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Pallavi Varshney
- Human Bioenergetics Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Sophia J Ghayur
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Bahadoran Z, Mirmiran P, Ghasemi A. Adipose organ dysfunction and type 2 diabetes: Role of nitric oxide. Biochem Pharmacol 2024; 221:116043. [PMID: 38325496 DOI: 10.1016/j.bcp.2024.116043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/07/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Adipose organ, historically known as specialized lipid-handling tissue serving as the long-term fat depot, is now appreciated as the largest endocrine organ composed of two main compartments, i.e., subcutaneous and visceral adipose tissue (AT), madding up white and beige/brown adipocytes. Adipose organ dysfunction manifested as maldistribution of the compartments, hypertrophic, hypoxic, inflamed, and insulin-resistant AT, contributes to the development of type 2 diabetes (T2D). Here, we highlight the role of nitric oxide (NO·) in AT (dys)function in relation to developing T2D. The key aspects determining lipid and glucose homeostasis in AT depend on the physiological levels of the NO· produced via endothelial NO· synthases (eNOS). In addition to decreased NO· bioavailability (via decreased expression/activity of eNOS or scavenging NO·), excessive NO· produced by inducible NOS (iNOS) in response to hypoxia and AT inflammation may be a critical interfering factor diverting NO· signaling to the formation of reactive oxygen and nitrogen species, resulting in AT and whole-body metabolic dysfunction. Pharmacological approaches boosting AT-NO· availability at physiological levels (by increasing NO· production and its stability), as well as suppression of iNOS-NO· synthesis, are potential candidates for developing NO·-based therapeutics in T2D.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Franco C, Canzoniero LMT. Zinc homeostasis and redox alterations in obesity. Front Endocrinol (Lausanne) 2024; 14:1273177. [PMID: 38260166 PMCID: PMC10800374 DOI: 10.3389/fendo.2023.1273177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Impairment of both cellular zinc and redox homeostasis is a feature of several chronic diseases, including obesity. A significant two-way interaction exists between redox metabolism and the relatively redox-inert zinc ion. Redox metabolism critically influences zinc homeostasis and controls its cellular availability for various cellular functions by regulating zinc exchange from/to zinc-binding proteins. Zinc can regulate redox metabolism and exhibits multiple pro-antioxidant properties. On the other hand, even minor disturbances in zinc status and zinc homeostasis affect systemic and cellular redox homeostasis. At the cellular level, zinc homeostasis is regulated by a multi-layered machinery consisting of zinc-binding molecules, zinc sensors, and two selective families of zinc transporters, the Zinc Transporter (ZnT) and Zrt, Irt-like protein (ZIP). In the present review, we summarize the current state of knowledge on the role of the mutual interaction between zinc and redox homeostasis in physiology and pathophysiology, pointing to the role of zinc in the alterations responsible for redox stress in obesity. Since zinc transporters primarily control zinc homeostasis, we describe how changes in the expression and activity of these zinc-regulating proteins are associated with obesity.
Collapse
|
4
|
Ting R, Dutton H, Sorisky A. In vitro studies of the renin-angiotensin system in human adipose tissue/adipocytes and possible relationship to SARS-CoV-2: a scoping review. Adipocyte 2023; 12:2194034. [PMID: 36973648 PMCID: PMC10054178 DOI: 10.1080/21623945.2023.2194034] [Citation(s) in RCA: 3] [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: 09/27/2022] [Accepted: 01/02/2023] [Indexed: 03/29/2023] Open
Abstract
The renin-angiotensin system (RAS) operates within adipose tissue. Obesity-related changes can affect adipose RAS, predisposing to hypertension, type 2 diabetes, and possibly severe COVID-19. We evaluated the in vitro research on human adipose RAS and identified gaps in the literature. Medline (Ovid), Embase (Ovid), Web of Science, Scopus, and 1findr were searched to identify relevant studies. Fifty primary studies met our inclusion criteria for analysis. Expression of RAS components (n = 14), role in differentiation (n = 14), association with inflammation (n = 15) or blood pressure (n = 7) were investigated. We found (1) obesity-related changes in RAS were frequently studied (30%); (2) an upswing of articles investigating adipose ACE-2 expression since the COVID-19 pandemic; (3) a paucity of papers on AT2R and Ang (1-7)/MasR which counterbalance Ang II/ART1; (4) weight loss lowered adipose ACE-2 mRNA expression; and (5) angiotensin receptor blockers (ARBs) reduced deleterious effects of angiotensin II. Overall, these studies link Ang II/ATR1 signalling to impaired adipogenesis and a pro-inflammatory dysfunctional adipose tissue, with ATR1 blockade limiting these responses. ACE-2 may mitigate Ang II effects by converting it to Ang(1-7) which binds MasR. More work is needed to understand adipose RAS in various pathologic states such as obesity and COVID-19 infection.T.
Collapse
Affiliation(s)
- Ryan Ting
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Heidi Dutton
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Department of Medicine, University of Ottawa, Ottawa, Canada
- The Ottawa Hospital/Ottawa Hospital Research Institute, Ottawa, Canada
| | - Alexander Sorisky
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Department of Medicine, University of Ottawa, Ottawa, Canada
- The Ottawa Hospital/Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
5
|
Sokolowski DJ, Vasquez OE, Wilson MD, Sokolowski MB, Anreiter I. Transcriptomic effects of the foraging gene shed light on pathways of pleiotropy and plasticity. Ann N Y Acad Sci 2023; 1526:99-113. [PMID: 37350250 DOI: 10.1111/nyas.15015] [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] [Indexed: 06/24/2023]
Abstract
Genes are often pleiotropic and plastic in their expression, features which increase and diversify the functionality of the genome. The foraging (for) gene in Drosophila melanogaster is highly pleiotropic and a long-standing model for studying individual differences in behavior and plasticity from ethological, evolutionary, and genetic perspectives. Its pleiotropy is known to be linked to its complex molecular structure; however, the downstream pathways and interactors remain mostly elusive. To uncover these pathways and interactors and gain a better understanding of how pleiotropy and plasticity are achieved at the molecular level, we explore the effects of different for alleles on gene expression at baseline and in response to 4 h of food deprivation, using RNA sequencing analysis in different Drosophila larval tissues. The results show tissue-specific transcriptomic dynamics influenced by for allelic variation and food deprivation, as well as genotype by treatment interactions. Differentially expressed genes yielded pathways linked to previously described for phenotypes and several potentially novel phenotypes. Together, these findings provide putative genes and pathways through which for might regulate its varied phenotypes in a pleiotropic, plastic, and gene-structure-dependent manner.
Collapse
Affiliation(s)
- Dustin J Sokolowski
- Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Oscar E Vasquez
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Michael D Wilson
- Genetics and Genome Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marla B Sokolowski
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
- Program in Child and Brain Development, Canadian Institute for Advanced Research, Toronto, Ontario, Canada
| | - Ina Anreiter
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Wang C, Wang X, Hu W. Molecular and cellular regulation of thermogenic fat. Front Endocrinol (Lausanne) 2023; 14:1215772. [PMID: 37465124 PMCID: PMC10351381 DOI: 10.3389/fendo.2023.1215772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Thermogenic fat, consisting of brown and beige adipocytes, dissipates energy in the form of heat, in contrast to the characteristics of white adipocytes that store energy. Increasing energy expenditure by activating brown adipocytes or inducing beige adipocytes is a potential therapeutic strategy for treating obesity and type 2 diabetes. Thus, a better understanding of the underlying mechanisms of thermogenesis provides novel therapeutic interventions for metabolic diseases. In this review, we summarize the recent advances in the molecular regulation of thermogenesis, focusing on transcription factors, epigenetic regulators, metabolites, and non-coding RNAs. We further discuss the intercellular and inter-organ crosstalk that regulate thermogenesis, considering the heterogeneity and complex tissue microenvironment of thermogenic fat.
Collapse
Affiliation(s)
- Cuihua Wang
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangdong, China
| | - Xianju Wang
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Wenxiang Hu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Bagheripour F, Jeddi S, Kashfi K, Ghasemi A. Metabolic effects of L-citrulline in type 2 diabetes. Acta Physiol (Oxf) 2023; 237:e13937. [PMID: 36645144 DOI: 10.1111/apha.13937] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/01/2023] [Accepted: 01/10/2023] [Indexed: 01/17/2023]
Abstract
The prevalence of type 2 diabetes (T2D) is increasing worldwide. Decreased nitric oxide (NO) bioavailability is involved in the pathophysiology of T2D and its complications. L-citrulline (Cit), a precursor of NO production, has been suggested as a novel therapeutic agent for T2D. Available data from human and animal studies indicate that Cit supplementation in T2D increases circulating levels of Cit and L-arginine while decreasing circulating glucose and free fatty acids and improving dyslipidemia. The underlying mechanisms for these beneficial effects of Cit include increased insulin secretion from the pancreatic β cells, increased glucose uptake by the skeletal muscle, as well as increased lipolysis and β-oxidation, and decreased glyceroneogenesis in the adipose tissue. Thus, Cit has antihyperglycemic, antidyslipidemic, and antioxidant effects and has the potential to be used as a new therapeutic agent in the management of T2D. This review summarizes available literature from human and animal studies to explore the effects of Cit on metabolic parameters in T2D. It also discusses the possible mechanisms underlying Cit-induced improved metabolic parameters in T2D.
Collapse
Affiliation(s)
- Fatemeh Bagheripour
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Johnston EK, Abbott RD. Adipose Tissue Paracrine-, Autocrine-, and Matrix-Dependent Signaling during the Development and Progression of Obesity. Cells 2023; 12:407. [PMID: 36766750 PMCID: PMC9913478 DOI: 10.3390/cells12030407] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Obesity is an ever-increasing phenomenon, with 42% of Americans being considered obese (BMI ≥ 30) and 9.2% being considered morbidly obese (BMI ≥ 40) as of 2016. With obesity being characterized by an abundance of adipose tissue expansion, abnormal tissue remodeling is a typical consequence. Importantly, this pathological tissue expansion is associated with many alterations in the cellular populations and phenotypes within the tissue, lending to cellular, paracrine, mechanical, and metabolic alterations that have local and systemic effects, including diabetes and cardiovascular disease. In particular, vascular dynamics shift during the progression of obesity, providing signaling cues that drive metabolic dysfunction. In this review, paracrine-, autocrine-, and matrix-dependent signaling between adipocytes and endothelial cells is discussed in the context of the development and progression of obesity and its consequential diseases, including adipose fibrosis, diabetes, and cardiovascular disease.
Collapse
Affiliation(s)
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
9
|
de Alwis N, Binder NK, Mangwiro YTM, Beard S, Pritchard N, Kadife E, Fato BR, Keenan E, Brownfoot FC, Kaitu’u-Lino TJ, Hannan NJ. Actions of Esomeprazole on the Maternal Vasculature in Lean and Obese Pregnant Mice with Impaired Nitric Oxide Synthesis: A Model of Preeclampsia. Int J Mol Sci 2022; 23:8185. [PMID: 35897759 PMCID: PMC9330120 DOI: 10.3390/ijms23158185] [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: 07/05/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 01/09/2023] Open
Abstract
Preeclampsia is a devastating, multisystem disorder of pregnancy. It has no cure except delivery, which if premature can impart significant neonatal morbidity. Efforts to repurpose pregnancy-safe therapeutics for the treatment of preeclampsia have led to the assessment of the proton pump inhibitor, esomeprazole. Preclinically, esomeprazole reduced placental secretion of anti-angiogenic sFlt-1, improved endothelial dysfunction, promoted vasorelaxation, and reduced maternal hypertension in a mouse model. Our understanding of the precise mechanisms through which esomeprazole works to reduce endothelial dysfunction and enhance vasoreactivity is limited. Evidence from earlier studies suggested esomeprazole might work via the nitric oxide pathway, upregulating endothelial nitric oxide synthase (eNOS). Here, we investigated the effect of esomeprazole in a mouse model of L-NAME-induced hypertension (decreased eNOS activity). We further antagonised the model by addition of diet-induced obesity, which is relevant to both preeclampsia and the nitric oxide pathway. Esomeprazole did not decrease blood pressure in this model, nor were there any alterations in vasoreactivity or changes in foetal outcomes in lean mice. We observed similar findings in the obese mouse cohort, except esomeprazole treatment enhanced ex vivo acetylcholine-induced vasorelaxation. As acetylcholine induces nitric oxide production, these findings hint at a function for esomeprazole in the nitric oxide pathway.
Collapse
Affiliation(s)
- Natasha de Alwis
- Therapeutics Discovery & Vascular Function Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.d.A.); (N.K.B.); (Y.T.M.M.); (S.B.); (B.R.F.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
| | - Natalie K. Binder
- Therapeutics Discovery & Vascular Function Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.d.A.); (N.K.B.); (Y.T.M.M.); (S.B.); (B.R.F.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
| | - Yeukai T. M. Mangwiro
- Therapeutics Discovery & Vascular Function Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.d.A.); (N.K.B.); (Y.T.M.M.); (S.B.); (B.R.F.)
| | - Sally Beard
- Therapeutics Discovery & Vascular Function Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.d.A.); (N.K.B.); (Y.T.M.M.); (S.B.); (B.R.F.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
| | - Natasha Pritchard
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
| | - Elif Kadife
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
- Obstetrics Diagnostics and Therapeutics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Bianca R. Fato
- Therapeutics Discovery & Vascular Function Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.d.A.); (N.K.B.); (Y.T.M.M.); (S.B.); (B.R.F.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
| | - Emerson Keenan
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
- Obstetrics Diagnostics and Therapeutics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Fiona C. Brownfoot
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
- Obstetrics Diagnostics and Therapeutics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Tu’uhevaha J. Kaitu’u-Lino
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
- Diagnostics Discovery and Reverse Translation in Pregnancy, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Natalie J. Hannan
- Therapeutics Discovery & Vascular Function Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.d.A.); (N.K.B.); (Y.T.M.M.); (S.B.); (B.R.F.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia; (N.P.); (E.K.); (E.K.); (F.C.B.); (T.J.K.-L.)
| |
Collapse
|
10
|
Chen CW, Chen LK, Huang TY, Yang DM, Liu SY, Tsai PJ, Chen TH, Lin HF, Juan CC. Nitric Oxide Mobilizes Intracellular Zn2+ via the GC/cGMP/PKG Signaling Pathway and Stimulates Adipocyte Differentiation. Int J Mol Sci 2022; 23:ijms23105488. [PMID: 35628299 PMCID: PMC9143299 DOI: 10.3390/ijms23105488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/03/2022] [Accepted: 05/11/2022] [Indexed: 12/10/2022] Open
Abstract
Plasma and tissue zinc ion levels are associated with the development of obesity. Previous studies have suggested that zinc ions may regulate adipocyte metabolism and that nitric oxide (NO) plays a pivotal role in the regulation of adipocyte physiology. Our previous study showed that chronic NO deficiency causes a significant decrease in adipose tissue mass in rats. Studies also suggested that zinc ions play an important modulatory role in regulating NO function. This study aims to explore the role of zinc ions in NO-regulated adipocyte differentiation. We hypothesized that NO could increase intracellular Zn2+ level and then stimulate adipocyte differentiation. ZnCl2 and the NO donor, NONOate, were used to explore the effects of Zn2+ and NO on adipocyte differentiation. Regulatory mechanisms of NO on intracellular Zn2+ mobilization were determined by detection. Then, Zn2+-selective chelator TPEN was used to clarify the role of intracellular Zn2+ on NO-regulated adipocyte differentiation. Furthermore, the relationship between adipocyte size, Zn2+ level, and NOS expression in human subcutaneous fat tissue was elucidated. Results showed that both ZnCl2 and NO stimulated adipocyte differentiation in a dose-dependent manner. NO stimulated intracellular Zn2+ mobilization in adipocytes through the guanylate cyclase (GC)/cyclic guanosine monophosphate (cGMP)/protein kinase G (PKG) pathway, and NO-stimulated adipocyte differentiation was Zn2+-dependent. In human subcutaneous adipose tissue, adipocyte size was negatively correlated with expression of eNOS. In conclusion, NO treatment stimulates intracellular Zn2+ mobilization through the GC/cGMP/PKG pathway, subsequently stimulating adipocyte differentiation.
Collapse
Affiliation(s)
- Chien-Wei Chen
- College of Human Development and Health, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan;
| | - Luen-Kui Chen
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
| | - Tai-Ying Huang
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
| | - De-Ming Yang
- Institute of Biophotonics, College of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan;
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Shui-Yu Liu
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
| | - Pei-Jiun Tsai
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (P.-J.T.); (T.-H.C.)
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Trauma Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Tien-Hua Chen
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (P.-J.T.); (T.-H.C.)
- Trauma Center, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Heng-Fu Lin
- Division of Trauma, Department of Surgery, Far-Eastern Memorial Hospital, New Taipei City 220216, Taiwan
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan 320315, Taiwan
- Correspondence: (H.-F.L.); (C.-C.J.)
| | - Chi-Chang Juan
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (L.-K.C.); (T.-Y.H.); (S.-Y.L.)
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Department of Education and Research, Taipei City Hospital, Taipei 103212, Taiwan
- Correspondence: (H.-F.L.); (C.-C.J.)
| |
Collapse
|
11
|
Ko H, Kim C, Lee MS, Chang E, Kim CT, Kim Y. High Hydrostatic Pressure Extract of Mulberry Leaf Attenuated Obesity-Induced Inflammation in Rats. J Med Food 2022; 25:251-260. [PMID: 35320014 DOI: 10.1089/jmf.2021.k.0113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Low-grade inflammation might be a link between obesity and obesity-associated metabolic dysfunction, including diabetes, hepatic steatosis, and other health complications. This study investigated whether the supplementation of high hydrostatic pressure extract of mulberry (Morus alba L.) leaves (HML) to obese rats could counteract obesity-related inflammation. Three-week-old male Sprague-Dawley rats were separated into three groups as follows: (a) a normal diet, (b) 45% high-fat (HF) diet, and HF diet containing 0.4% HML (c) or 0.8% HML (d) (IACUC No. 17-033). After 14 weeks of HML supplementation, adipose tissue mass, mRNA expression of adipogenic genes, such as aP2, peroxisome proliferator-activated receptor γ (PPARγ), and sterol regulatory element binding protein 1c (SREBP1c), and macrophage recruitment were significantly decreased in HF-fed obese rats. Serum concentrations of nitric oxide and mRNA levels of arginase1 (Arg1), CD11c, and inducible nitric oxide synthase (iNOS) involved in adipose tissue macrophage M1 polarization were also significantly reduced by HML. Moreover, HML alleviated the serum and hepatic lipid profiles and reduced hepatic lipogenic gene expression of acetyl-CoA carboxylase (ACC), cluster of differentiation 36 (CD36), CPT1, fatty acid synthase (FAS), stearoyl-CoA desaturase (SCD1), and SREBP1c, and inflammation-associated genes, including IL1β, interleukin 6 (IL6), and tumor necrosis factor α (TNFα). Serum IL6 and TNFα levels were remarkedly suppressed in the 0.8% HML group. These results suggested that the favorable effect of HML on obesity-associated inflammation might be related in part to the decrease in adipose tissue and hepatic fat deposition and inflammation.
Collapse
Affiliation(s)
- Hyunmi Ko
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Chaemin Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Mak-Soon Lee
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea
| | - Eugene Chang
- Department of Food and Nutrition, Gangneung-Wonju National University, Gangneung-si, Korea
| | | | - Yangha Kim
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Korea.,Graduate Program in System Health Science and Engineering, Ewha Womans University, Seoul, Korea
| |
Collapse
|
12
|
Sebag SC, Zhang Z, Qian Q, Li M, Zhu Z, Harata M, Li W, Zingman LV, Liu L, Lira VA, Potthoff MJ, Bartelt A, Yang L. ADH5-mediated NO bioactivity maintains metabolic homeostasis in brown adipose tissue. Cell Rep 2021; 37:110003. [PMID: 34788615 PMCID: PMC8640996 DOI: 10.1016/j.celrep.2021.110003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/23/2021] [Accepted: 10/22/2021] [Indexed: 01/21/2023] Open
Abstract
Brown adipose tissue (BAT) thermogenic activity is tightly regulated by cellular redox status, but the underlying molecular mechanisms are incompletely understood. Protein S-nitrosylation, the nitric-oxide-mediated cysteine thiol protein modification, plays important roles in cellular redox regulation. Here we show that diet-induced obesity (DIO) and acute cold exposure elevate BAT protein S-nitrosylation, including UCP1. This thermogenic-induced nitric oxide bioactivity is regulated by S-nitrosoglutathione reductase (GSNOR; alcohol dehydrogenase 5 [ADH5]), a denitrosylase that balances the intracellular nitroso-redox status. Loss of ADH5 in BAT impairs cold-induced UCP1-dependent thermogenesis and worsens obesity-associated metabolic dysfunction. Mechanistically, we demonstrate that Adh5 expression is induced by the transcription factor heat shock factor 1 (HSF1), and administration of an HSF1 activator to BAT of DIO mice increases Adh5 expression and significantly improves UCP1-mediated respiration. Together, these data indicate that ADH5 controls BAT nitroso-redox homeostasis to regulate adipose thermogenesis, which may be therapeutically targeted to improve metabolic health.
Collapse
Affiliation(s)
- Sara C. Sebag
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,These authors contributed equally
| | - Zeyuan Zhang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,These authors contributed equally
| | - Qingwen Qian
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mark Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Zhiyong Zhu
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mikako Harata
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Wenxian Li
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Leonid V. Zingman
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, Cardiovascular Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Limin Liu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Vitor A. Lira
- Department of Health and Human Physiology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J. Potthoff
- Department of Neuroscience and Pharmacology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention, Ludwig Maximilians University Munich Pettenkoferstr. 9, 80336 Munich, Germany,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Technische Universität München, Biedersteiner Str. 29, 80802 München, Germany,Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Ingolstädter Landstr. 1, 85764 Neuherberg, Germany,Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA,Lead contact,Correspondence:
| |
Collapse
|
13
|
Qin A, Chen S, Wang P, Huang X, Zhang Y, Liang L, Du LR, Lai DH, Ding L, Yu X, Xiang AP. Knockout of NOS2 Promotes Adipogenic Differentiation of Rat MSCs by Enhancing Activation of JAK/STAT3 Signaling. Front Cell Dev Biol 2021; 9:638518. [PMID: 33816486 PMCID: PMC8017136 DOI: 10.3389/fcell.2021.638518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogeneous population of cells that possess multilineage differentiation potential and extensive immunomodulatory properties. In mice and rats, MSCs produce nitric oxide (NO), as immunomodulatory effector molecule that exerts an antiproliferative effect on T cells, while the role of NO in differentiation was less clear. Here, we investigated the role of NO synthase 2 (NOS2) on adipogenic and osteogenic differentiation of rat MSCs. MSCs isolated from NOS2-null (NOS2–/–) and wild type (WT) Sprague–Dawley (SD) rats exhibited homogenous fibroblast-like morphology and characteristic phenotypes. However, after induction, adipogenic differentiation was found significantly promoted in NOS2–/– MSCs compared to WT MSCs, but not in osteogenic differentiation. Accordingly, qRT-PCR revealed that the adipogenesis-related genes PPAR-γ, C/EBP-α, LPL and FABP4 were markedly upregulated in NOS2–/– MSCs, but not for osteogenic transcription factors or marker genes. Further investigations revealed that the significant enhancement of adipogenic differentiation in NOS2–/– MSCs was due to overactivation of the STAT3 signaling pathway. Both AG490 and S3I-201, small molecule inhibitors that selectively inhibit STAT3 activation, reversed this adipogenic effect. Furthermore, after high-fat diet (HFD) feeding, knockout of NOS2 in rat MSCs resulted in significant obesity. In summary, NOS2 is involved in the regulation of rat MSC adipogenic differentiation via the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Aiping Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Sheng Chen
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ping Wang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiaotao Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lu Liang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ling-Ran Du
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - De-Hua Lai
- Center for Parasitic Organisms, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Li Ding
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Porwal K, Pal S, Bhagwati S, Siddiqi MI, Chattopadhyay N. Therapeutic potential of phosphodiesterase inhibitors in the treatment of osteoporosis: Scopes for therapeutic repurposing and discovery of new oral osteoanabolic drugs. Eur J Pharmacol 2021; 899:174015. [PMID: 33711307 DOI: 10.1016/j.ejphar.2021.174015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/19/2021] [Accepted: 03/03/2021] [Indexed: 01/05/2023]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are ubiquitously expressed enzymes that hydrolyze phosphodiester bond in the second messenger molecules including cAMP and cGMP. A wide range of drugs blocks one or more PDEs thereby preventing the inactivation of cAMP/cGMP. PDEs are differentially expressed in bone cells including osteoblasts, osteoclasts and chondrocytes. Intracellular increases in cAMP/cGMP levels in osteoblasts result in osteogenic response. Acting via the type 1 PTH receptor, teriparatide and abaloparatide increase intracellular cAMP and induce osteoanabolic effect, and many PDE inhibitors mimic this effect in preclinical studies. Since all osteoanabolic drugs are injectable and that oral drugs are considered to improve the treatment adherence and persistence, osteogenic PDE inhibitors could be a promising alternative to the currently available osteogenic therapies and directly assessed clinically in drug repurposing mode. Similar to teriparatide/abaloparatide, PDE inhibitors while stimulating osteoblast function also promote osteoclast function through stimulation of receptor activator of nuclear factor kappa-B ligand production from osteoblasts. In this review, we critically discussed the effects of PDE inhibitors in bone cells from cellular signalling to a variety of preclinical models that evaluated the bone formation mechanisms. We identified pentoxifylline (a non-selective PDE inhibitor) and rolipram (a PDE4 selective inhibitor) being the most studied inhibitors with osteogenic effect in preclinical models of bone loss at ≤ human equivalent doses, which suggest their potential for post-menopausal osteoporosis treatment through therapeutic repurposing. Subsequently, we treated pentoxifylline and rolipram as prototypical osteogenic PDEs to predict new chemotypes via the computer-aided design strategies for new drugs, based on the structural biology of PDEs.
Collapse
Affiliation(s)
- Konica Porwal
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), India
| | - Subhashis Pal
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), India
| | - Sudha Bhagwati
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Sector 10/1 Jankipuram Extension, Sitapur Road, Lucknow, 226 031, India
| | - Mohd Imran Siddiqi
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Sector 10/1 Jankipuram Extension, Sitapur Road, Lucknow, 226 031, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Centre for Research in Anabolic Skeletal Targets in Health and Illness (ASTHI), India.
| |
Collapse
|
15
|
Protein Digests and Pure Peptides from Chia Seed Prevented Adipogenesis and Inflammation by Inhibiting PPARγ and NF-κB Pathways in 3T3L-1 Adipocytes. Nutrients 2021; 13:nu13010176. [PMID: 33430086 PMCID: PMC7826547 DOI: 10.3390/nu13010176] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/25/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022] Open
Abstract
The objective was to evaluate the mechanisms of digested total proteins (DTP), albumin, glutelin, and pure peptides from chia seed (Salvia hispanica L.) to prevent adipogenesis and its associated inflammation in 3T3-L1 adipocytes. Preadipocytes (3T3-L1) were treated during differentiation with either DTP or digested albumin or glutelin (1 mg/mL) or pure peptides NSPGPHDVALDQ and RMVLPEYELLYE (100 µM). Differentiated adipocytes also received DTP, digested albumin or glutelin (1 mg/mL), before (prevention) or after (inhibition) induced inflammation by addition of conditioned medium (CM) from inflamed macrophages. All treatments prevented adipogenesis, reducing more than 50% the expression of PPARγ and to a lesser extent lipoprotein lipase (LPL), fatty acid synthase (FAS), sterol regulatory element-binding protein 1 (SREBP1), lipase activity and triglycerides. Inflammation induced by CM was reduced mainly during prevention, while DTP decreased expression of NF-κB (−48.4%), inducible nitric oxide synthase (iNOS) (−46.2%) and COX-2 (−64.5%), p < 0.05. Secretions of nitric oxide, PGE2 and TNFα were reduced by all treatments, p < 0.05. DTP reduced expressions of iNOS (−52.1%) and COX-2 (−66.4%). Furthermore, digested samples and pure peptides prevented adipogenesis by modulating PPARγ and additionally, preventing and even inhibiting inflammation in adipocytes by inhibition of PPARγ and NF-κB expression. These results highlight the effectiveness of digested total proteins and peptides from chia seed against adipogenesis complications in vitro.
Collapse
|
16
|
Chinnadurai R, Ibrahim ST, Tay T, Bhutani S, Kalra PA. Body weight-based initial dosing of tacrolimus in renal transplantation: Is this an ideal approach? J Ren Care 2020; 47:51-57. [PMID: 32730692 DOI: 10.1111/jorc.12339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/04/2020] [Accepted: 03/15/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Tacrolimus dosing immediately posttransplant is based on body weight. Recent studies have highlighted that the dosing of tacrolimus purely based on weight may not be appropriate, particularly in individuals who are obese. OBJECTIVES This study aimed to estimate the effect of body mass index (BMI) and the weight-based dosing on tacrolimus trough levels in recipients of renal transplants. DESIGN AND PARTICIPANTS This study was conducted on 400 of the 863 patients registered in the Salford, UK, renal transplant database between 2012 and 2019 who had complete and analysable datasets. Data were collected at baseline (first tacrolimus trough level after transplantation), after 1 month and 6 months posttransplantation. The cohort was split into three groups based on BMI (kg/m2 ; Group 1 ≤ 25, Group 2 > 25-30 and Group 3 > 30) which were compared with respect to tacrolimus dose, plasma levels and concentration/dose (C/D) ratio at the three-time points. RESULTS Patients in the higher BMI group (Group 3) had significantly higher baseline tacrolimus trough levels despite receiving a lower initiation dose per kilogram of body weight. After 1 and 6-months posttransplant, the higher BMI group were receiving a significantly lower tacrolimus dose relative to their body weight, with a significant negative correlation between body weight and tacrolimus/kg body weight. There was no adverse relationship evident between tacrolimus dosing or concentration and graft function. CONCLUSIONS Our study showed that standard dosing of tacrolimus based on body weight in individuals who were obese did not adversely affect their tacrolimus concentrations or transplant function.
Collapse
Affiliation(s)
- Rajkumar Chinnadurai
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK.,Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Sara T Ibrahim
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK.,Department of Internal Medicine and Nephrology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Tricia Tay
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| | - Shiv Bhutani
- Manchester Institute of Nephrology and Transplantation, Manchester University NHS Foundation Trust, Manchester, UK
| | - Philip A Kalra
- Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK.,Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, UK
| |
Collapse
|
17
|
Chung KH, Chiou HY, Chang JS, Chen YH. Associations of nitric oxide with obesity and psychological traits among children and adolescents in Taiwan. Pediatr Obes 2020; 15:e12593. [PMID: 31786830 DOI: 10.1111/ijpo.12593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 09/29/2019] [Accepted: 10/11/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Findings concerning nitric oxide (NO) in children and adolescents with obesity are scant. OBJECTIVE This study examined the links of NO with obesity and psychological traits (ie, self-concept, anxiety, depression, anger and disruptive behaviour) in children and adolescents in Taiwan. METHODS A total of 564 first, fourth and seventh graders (314 children with overweight/obesity and 250 children with normal weight) completed an in-hospital health examination in 2010. All students received a physical examination, underwent blood sample collection and completed a questionnaire. Multiple linear regression analyses were performed for analyses. RESULTS Among the fourth and seventh graders (P=.003 and.001, respectively), the students with overweight/obesity displayed significantly higher levels of NO than those with normal weight; however, no difference was observed in males and females. In multiple linear regression models, a high level of anxiety was independently associated with low NO levels (β=-1.33, 95% confidence interval -2.24 to -0.41) in first graders who with overweight/obesity. No association between NO levels and psychological traits was evident among students with normal weight. CONCLUSIONS Our results enrich the limited data and suggest that NO may be associated with obesity and psychopathology and should be a concern in the pathophysiology of childhood mental health and obesity.
Collapse
Affiliation(s)
- Kuo-Hsuan Chung
- Department of Psychiatry and Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yi Chiou
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Jung-Su Chang
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan
| | - Yi-Hua Chen
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
18
|
Bahadoran Z, Mirmiran P, Ghasemi A. Role of Nitric Oxide in Insulin Secretion and Glucose Metabolism. Trends Endocrinol Metab 2020; 31:118-130. [PMID: 31690508 DOI: 10.1016/j.tem.2019.10.001] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 01/20/2023]
Abstract
Nitric oxide (NO) contributes to carbohydrate metabolism and decreased NO bioavailability is involved in the development of type 2 diabetes mellitus (T2DM). NO donors may improve insulin signaling and glucose homeostasis in T2DM and insulin resistance (IR), suggesting the potential clinical importance of NO-based interventions. In this review, site-specific roles of the NO synthase (NOS)-NO pathway in carbohydrate metabolism are discussed. In addition, the metabolic effects of physiological low levels of NO produced by constitutive NOS (cNOS) versus pathological high levels of NO produced by inducible NOS (iNOS) in pancreatic β-cells, adipocytes, hepatocytes, and skeletal muscle cells are summarized. A better understanding of the NOS-NO system in the regulation of glucose homeostasis can hopefully facilitate the development of new treatments for T2DM.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Human Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Gheibi S, Samsonov AP, Gheibi S, Vazquez AB, Kashfi K. Regulation of carbohydrate metabolism by nitric oxide and hydrogen sulfide: Implications in diabetes. Biochem Pharmacol 2020; 176:113819. [PMID: 31972170 DOI: 10.1016/j.bcp.2020.113819] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/15/2020] [Indexed: 12/19/2022]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are two gasotransmitters that are produced in the human body and have a key role in many of the physiological activities of the various organ systems. Decreased NO bioavailability and deficiency of H2S are involved in the pathophysiology of type 2 diabetes and its complications. Restoration of NO levels have favorable metabolic effects in diabetes. The role of H2S in pathophysiology of diabetes is however controversial; H2S production is decreased during development of obesity, diabetes, and its complications, suggesting the potential therapeutic effects of H2S. On the other hand, increased H2S levels disturb the pancreatic β-cell function and decrease insulin secretion. In addition, there appear to be important interactions between NO and H2S at the levels of both biosynthesis and signaling pathways, yet clear an insight into this relationship is lacking. H2S potentiates the effects of NO in the cardiovascular system as well as NO release from its storage pools. Likewise, NO increases the activity and the expression of H2S-generating enzymes. Inhibition of NO production leads to elimination/attenuation of the cardioprotective effects of H2S. Regarding the increasing interest in the therapeutic applications of NO or H2S-releasing molecules in a variety of diseases, particularly in the cardiovascular disorders, much is to be learned about their function in glucose/insulin metabolism, especially in diabetes. The aim of this review is to provide a better understanding of the individual and the interactive roles of NO and H2S in carbohydrate metabolism.
Collapse
Affiliation(s)
- Sevda Gheibi
- Department of Clinical Sciences in Malmö, Unit of Molecular Metabolism, Lund University Diabetes Centre, Clinical Research Center, Malmö University Hospital, Lund University, Malmö, Sweden.
| | - Alan P Samsonov
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Shahsanam Gheibi
- Maternal and Childhood Obesity Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Alexandra B Vazquez
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, NY, USA.
| |
Collapse
|
20
|
Foroumandi E, Alizadeh M, Kheirouri S, Asghari Jafarabadi M. Exploring the role of body mass index in relationship of serum nitric oxide and advanced glycation end products in apparently healthy subjects. PLoS One 2019; 14:e0213307. [PMID: 30856212 PMCID: PMC6411143 DOI: 10.1371/journal.pone.0213307] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/18/2019] [Indexed: 11/19/2022] Open
Abstract
This study aimed to identify any association of serum nitric oxide (NO) and advanced glycation end products (AGEs) with body mass index (BMI) in apparently healthy subjects. In this cross-sectional study, participants were 90 apparently healthy subjects, categorized into three BMI groups as follows: BMI≤19.5 (n = 21), 19.6≤BMI≤24.9 (n = 35), and BMI≥25 (n = 34). Serum levels of NO were measured by griess reaction method. Determination of serum pentosidine and carboxymethyllysine (CML) was done using ELISA. Median (95% confidence interval [CI]: lower- upper) of serum NO in subjects with BMI≥25 were 68.94 (CI: 55.01–70.56) μmol/L, which was higher compared with 19.6≤BMI≤24.9 and BMI≤19.5 groups (22.65 (CI: 19.29–28.17) μmol/L and 8.00 (CI: 9.12–29.58) μmol/L, respectively). Serum NO positively correlated with BMI in total subjects (r = 0.585, p<0.001), which this correlation was significant in both male and female groups (r = 0.735, p<0.001 and r = 0.476, p = 0.001, respectively). Serum pentosidine and CML were significantly lower in subjects with higher BMI. Further, BMI showed negative correlations with pentosidine and CML (r = -0.363, p<0.001 and r = -0.484, p<0.001, respectively). There were not any significant differences in serum NO, pentosidine, and CML levels between sex groups. After adjusting the effects of confounders (BMI, sex, age, and waist to hip ratio), serum NO significantly correlated with serum pentosidine and CML (r = -0.319, p = 0.003 and r = -0.433, p<0.001, respectively). It is concluded that higher BMI is accompanied by increased serum NO and suppressed pentosidine and CML.
Collapse
Affiliation(s)
- Elaheh Foroumandi
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Students’ Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Alizadeh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- * E-mail:
| | - Sorayya Kheirouri
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
21
|
Bahadoran Z, Mirmiran P, Jeddi S, Carlström M, Azizi F, Ghasemi A. Circulating markers of nitric oxide homeostasis and cardiometabolic diseases: insights from population-based studies. Free Radic Res 2019; 53:359-376. [PMID: 30821533 DOI: 10.1080/10715762.2019.1587168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Emerging data suggest that impaired nitric oxide (NO) homeostasis has a key role in development of cardiometabolic disorders. The association between circulating levels of NO metabolites, i.e. nitrate and nitrite (NOx), and risk of chronic diseases has not yet been fully clarified. This work aims to address epidemiologic aspects of NO metabolism and discusses different physiologic and pathophysiologic conditions influencing circulating NOx. Further, cross-sectional associations of serum NOx with metabolic disorders are described and along the way, potential short-term and long-term power of serum NOx for predicting cardiometabolic outcomes are reviewed. Results from population-based studies show that circulating NOx is affected by aging, smoking habits, pregnancy, menopause status, thyroid hormones, and various pathologic conditions including type 2 diabetes, insulin resistance, hypertension, and renal dysfunction. Lifestyle factors, especially dietary habits, but also smoking habits and the degree of physical activity influence NO homeostasis and the circulating levels of NOx. Elevated serum NOx, due to increased iNOS activity, is associated with increased incidence of metabolic syndrome, different obesity phenotypes, and cardiovascular events.
Collapse
Affiliation(s)
- Zahra Bahadoran
- a Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Parvin Mirmiran
- b Department of Clinical Nutrition and Diet Therapy, Faculty of Nutrition Sciences and Food Technology , National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Sajad Jeddi
- c Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mattias Carlström
- d Department of Physiology and Pharmacology , Karolinska Institutet , Stockholm , Sweden
| | - Fereidoun Azizi
- e Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Asghar Ghasemi
- c Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| |
Collapse
|
22
|
Atawia RT, Bunch KL, Toque HA, Caldwell RB, Caldwell RW. Mechanisms of obesity-induced metabolic and vascular dysfunctions. FRONT BIOSCI-LANDMRK 2019; 24:890-934. [PMID: 30844720 PMCID: PMC6689231 DOI: 10.2741/4758] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Obesity has reached epidemic proportions and its prevalence is climbing. Obesity is characterized by hypertrophied adipocytes with a dysregulated adipokine secretion profile, increased recruitment of inflammatory cells, and impaired metabolic homeostasis that eventually results in the development of systemic insulin resistance, a phenotype of type 2 diabetes. Nitric oxide synthase (NOS) is an enzyme that converts L-arginine to nitric oxide (NO), which functions to maintain vascular and adipocyte homeostasis. Arginase is a ureohydrolase enzyme that competes with NOS for L-arginine. Arginase activity/expression is upregulated in obesity, which results in diminished bioavailability of NO, impairing both adipocyte and vascular endothelial cell function. Given the emerging role of NO in the regulation of adipocyte physiology and metabolic capacity, this review explores the interplay between arginase and NO, and their effect on the development of metabolic disorders, cardiovascular diseases, and mitochondrial dysfunction in obesity. A comprehensive understanding of the mechanisms involved in the development of obesity-induced metabolic and vascular dysfunction is necessary for the identification of more effective and tailored therapeutic avenues for their prevention and treatment.
Collapse
Affiliation(s)
- Reem T Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Katharine L Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology,and Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904, USA
| | - Robert W Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University. Augusta, GA 30904,USA,
| |
Collapse
|
23
|
Aggarwal H, Kanuri BN, Dikshit M. Role of iNOS in Insulin Resistance and Endothelial Dysfunction. OXIDATIVE STRESS IN HEART DISEASES 2019:461-482. [DOI: 10.1007/978-981-13-8273-4_21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
24
|
Abstract
During the last decades, research on adipose tissues has spread in parallel with the extension of obesity. Several observations converged on the idea that adipose tissues are organized in a large organ with endocrine and plastic properties. Two parenchymal components: white (WATs) and brown adipose tissues (BATs) are contained in subcutaneous and visceral compartments. Although both have endocrine properties, their function differs: WAT store lipids to allow intervals between meals, BAT burns lipids for thermogenesis. In spite of these opposite functions, they share the ability for reciprocal reversible transdifferentiation to tackle special physiologic needs. Thus, chronic need for thermogenesis induces browning and chronic positive energy balance induce whitening. Lineage tracing and data from explant studies strongly suggest other remodeling properties of this organ. During pregnancy and lactation breast WAT transdifferentiates into milk-secreting glands, composed by cells with abundant cytoplasmic lipids (pink adipocytes) and in the postlactation period pink adipocytes transdifferentiate back into WAT and BAT. The plastic properties of mature adipocytes are supported also by a liposecretion process in vitro where adult cell in culture transdifferentiate to differentiated fibroblast-like elements able to give rise to different phenotypes (rainbow adipocytes). In addition, the inflammasome system is activated in stressed adipocytes from obese adipose tissue. These adipocytes die and debris are reabsorbed by macrophages inducing a chronic low-grade inflammation, potentially contributing to insulin resistance and T2 diabetes. Thus, the plastic properties of this organ could open new therapeutic perspectives in the obesity-related metabolic disease and in breast pathologies. © 2018 American Physiological Society. Compr Physiol 8:1357-1431, 2018.
Collapse
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
25
|
Bakhtiarzadeh F, Siavoshi F, Gheibi S, Kashfi K, Samadi R, Jeddi S, Ghasemi A. Effects of long-term oral nitrate administration on adiposity in normal adult female rats. Life Sci 2018; 210:76-85. [PMID: 30118772 DOI: 10.1016/j.lfs.2018.08.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/04/2018] [Accepted: 08/12/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Nitric oxide (NO) deficiency is associated with obesity. Nitrate could act as a substrate for production of NO and is a novel therapeutic agent in obesity. This study aims at determining effects of long-term nitrate administration on obesity indices in normal adult female rats. METHODS Female Wistar rats were divided into four groups (n = 10/each): i.e. control group received tap water and three treatment groups received water containing 50, 100 and 150 mg/L sodium nitrate for 6 months. Body weight (g) was measured monthly; naso-anal length (cm) and obesity indices including body mass index (BMI), Lee index, abdominal and thoracic circumferences were determined every two months. Both white adipose tissue (WAT) and brown adipose tissue (BAT) were weighted and then adiposity index was calculated. In addition, level of NOx (nitrate + nitrite) in serum and adipose tissues were measured at the end of the study. RESULTS Compared to controls, body weights and naso-anal length were significantly (P < 0.001) lower in all nitrate-treated rats. Compared to controls, nitrate-treated rats had also lower adiposity indices, BMI, Lee index, abdominal and thoracic circumferences (13%, 17% and 22% for BMI and 5%, 6% and 8% for lee index at dose 50, 100, and 150 mg/L, respectively). In addition, nitrate administration increased NOx levels in serum and adipose tissues. CONCLUSIONS Long-term nitrate administration has favorable effects on adiposity. It increases brown and decreases white adipose tissues in normal female rats; these observations could potentially help in management of obesity.
Collapse
Affiliation(s)
- Fatemeh Bakhtiarzadeh
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Siavoshi
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sevda Gheibi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, NY, USA
| | - Roghaieh Samadi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Allerton TD, Proctor DN, Stephens JM, Dugas TR, Spielmann G, Irving BA. l-Citrulline Supplementation: Impact on Cardiometabolic Health. Nutrients 2018; 10:nu10070921. [PMID: 30029482 PMCID: PMC6073798 DOI: 10.3390/nu10070921] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022] Open
Abstract
Diminished bioavailability of nitric oxide (NO), the gaseous signaling molecule involved in the regulation of numerous vital biological functions, contributes to the development and progression of multiple age- and lifestyle-related diseases. While l-arginine is the precursor for the synthesis of NO by endothelial-nitric oxide synthase (eNOS), oral l-arginine supplementation is largely ineffective at increasing NO synthesis and/or bioavailability for a variety of reasons. l-citrulline, found in high concentrations in watermelon, is a neutral alpha-amino acid formed by enzymes in the mitochondria that also serves as a substrate for recycling l-arginine. Unlike l-arginine, l-citrulline is not quantitatively extracted from the gastrointestinal tract (i.e., enterocytes) or liver and its supplementation is therefore more effective at increasing l-arginine levels and NO synthesis. Supplementation with l-citrulline has shown promise as a blood pressure lowering intervention (both resting and stress-induced) in adults with pre-/hypertension, with pre-clinical (animal) evidence for atherogenic-endothelial protection. Preliminary evidence is also available for l-citrulline-induced benefits to muscle and metabolic health (via vascular and non-vascular pathways) in susceptible/older populations. In this review, we examine the impact of supplementing this important urea cycle intermediate on cardiovascular and metabolic health outcomes and identify future directions for investigating its therapeutic impact on cardiometabolic health.
Collapse
Affiliation(s)
| | - David N Proctor
- Department of Kinesiology, Pennsylvania State University, University Park, PA 16802, USA.
| | | | - Tammy R Dugas
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Guillaume Spielmann
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA.
| | - Brian A Irving
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
27
|
Yang S, Guo L, Su Y, Wen J, Du J, Li X, Liu Y, Feng J, Xie Y, Bai Y, Wang H, Liu Y. Nitric oxide balances osteoblast and adipocyte lineage differentiation via the JNK/MAPK signaling pathway in periodontal ligament stem cells. Stem Cell Res Ther 2018; 9:118. [PMID: 29716662 PMCID: PMC5930947 DOI: 10.1186/s13287-018-0869-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/28/2018] [Accepted: 04/12/2018] [Indexed: 12/21/2022] Open
Abstract
Background Critical tissues that undergo regeneration in periodontal tissue are of mesenchymal origin; thus, investigating the regulatory mechanisms underlying the fate of periodontal ligament stem cells could be beneficial for application in periodontal tissue regeneration. Nitric oxide (NO) regulates many biological processes in developing embryos and adult stem cells. The present study was designed to investigate the effects of NO on the function of human periodontal ligament stem cells (PDLSCs) as well as to elucidate the underlying molecular mechanisms. Methods Immunofluorescent staining and flow cytometry were used for stem cell identification. Western blot, reverse transcription polymerase chain reaction (RT-PCR), immunofluorescent staining, and flow cytometry were used to examine the expression of NO-synthesizing enzymes. The proliferative capacity of PDLSCs was determined by EdU assays. The osteogenic potential of PDLSCs was tested using alkaline phosphatase (ALP) staining, Alizarin Red staining, and calcium concentration detection. Oil Red O staining was used to analyze the adipogenic ability. Western blot, RT-PCR, and staining were used to examine the signaling pathway. Results Human PDLSCs expressed both inducible NO synthase (iNOS) and endothelial NO synthase (eNOS) and produced NO. Blocking the generation of NO with the NOS inhibitor l-NG-monomethyl arginine (l-NMMA) had no influence on PDLSC proliferation and apoptosis but significantly attenuated the osteogenic differentiation capacity and stimulated the adipogenic differentiation capacity of PDLSCs. Increasing the physiological level of NO with NO donor sodium nitroprusside (SNP) significantly promoted the osteogenic differentiation capacity but reduced the adipogenic differentiation capacity of PDLSCs. NO balances the osteoblast and adipocyte lineage differentiation in periodontal ligament stem cells via the c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK) signaling pathway. Conclusions NO is essential for maintaining the balance between osteoblasts and adipocytes in PDLSCs via the JNK/MAPK signaling pathway. Graphical Abstract NO balances osteoblast and adipocyte lineage differentiation via JNK/MAPK signaling pathway![]() Electronic supplementary material The online version of this article (10.1186/s13287-018-0869-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shan Yang
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Lijia Guo
- Department of Orthodontics, Capital Medical University School of Stomatology, Beijing, People's Republic of China
| | - Yingying Su
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jing Wen
- Department of Orthodontics, Capital Medical University School of Stomatology, Beijing, People's Republic of China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Yitong Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Jie Feng
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Yongmei Xie
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China
| | - Yuxing Bai
- Department of Orthodontics, Capital Medical University School of Stomatology, Beijing, People's Republic of China
| | - Hao Wang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Tian Tan Xi Li No.4, Beijing, 100050, People's Republic of China.
| |
Collapse
|
28
|
Ekambaram P, Parasuraman P. Differential expression of sirtuin 2 and adipocyte maturation restriction: an adaptation process during hypoxia in fish. Biol Open 2017; 6:1375-1382. [PMID: 28808139 PMCID: PMC5612243 DOI: 10.1242/bio.027334] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Sirtuins have received widespread attention due to their diverse physiological role in metabolism. Among sirtuins, SIRT2 is more abundant in adipocytes and exerts effects on adipocyte differentiation, a process which involves conversion of preadipocytes to mature adipocytes orchestrated by adipokines and adipogenic transcription factors. Grey mullet (Mugil cephalus) was chosen as a study organism due to its excellent service as a biomonitor. Adipocytes isolated from natural field conditions were termed as field-hypoxic (Ennore) and -normoxic (Kovalam) based on dissolved oxygen (DO) level in the estuary. A previous study portrayed the hypoxic instance of Ennore estuary (low DO) and grey mullet [HIF1α in adipocytes, brain endothelial cell (EC) and hepatocytes] inhabiting this estuary (
Padmini et al., 2016a,
b; Padmini and Tharani, 2015). In this context, fish adipocytes of both conditions were subjected to in vitro hypoxia for 1 h (in the pre/trigassed incubator with the supply of 1% O2; 94% N2; 5% CO2) and were analysed for the expression of adipokines, adipogenic transcription factors and anti-adipogenic markers in fish adipocytes. Elevation of asymmetric dimethylarginine (ADMA), TNFα and leptin along with decreased adiponectin, adipogenic transcription factors and altering sirtuins were observed in test adipocytes and in control adipocytes on in vitro hypoxia. This suggests that adipocytes may follow internal caloric restriction as portrayed from cytomorphological/ultrastructural analysis, limiting adipocyte maturation process, one of the adaptive mechanisms triggered by adipocyte of fish surviving in Ennore estuary. Prolonged exposure to hypoxia (test on in vitro hypoxia for 1 h) showed a drastic alteration in these components leading to both structural and biological fluctuation when compared to limited hypoxic condition (field-hypoxic and control on in vitro hypoxia). Our study concludes that hypoxia may serve as the chief molecular cue in eliciting adipocyte maturation restriction though metabolic reprogramming and it also shows the significance of adipocyte maturation restriction in imparting survival mechanism. Summary: Adipocyte maturation restriction is tightly regulated by SIRT2 activation which downregulates preadipocytes from the maturation process as adaptation strategy in fish surviving in the polluted (hypoxic) environment.
Collapse
Affiliation(s)
- Padmini Ekambaram
- P.G. Department of Biochemistry, Bharathi Women's College, Affiliated to University of Madras, Tamil Nadu, Chennai-600 108, India
| | - Parimala Parasuraman
- P.G. Department of Biochemistry, Bharathi Women's College, Affiliated to University of Madras, Tamil Nadu, Chennai-600 108, India
| |
Collapse
|
29
|
Frigolet ME, Thomas G, Beard K, Lu H, Liu L, Fantus IG. The bradykinin-cGMP-PKG pathway augments insulin sensitivity via upregulation of MAPK phosphatase-5 and inhibition of JNK. Am J Physiol Endocrinol Metab 2017; 313:E321-E334. [PMID: 28679626 DOI: 10.1152/ajpendo.00298.2016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/13/2023]
Abstract
Bradykinin (BK) promotes insulin sensitivity and glucose uptake in adipocytes and other cell types. We demonstrated that in rat adipocytes BK enhances insulin-stimulated glucose transport via endothelial nitric oxide synthase, nitric oxide (NO) generation, and decreased activity of the mitogen-activated protein kinase (MAPK) JNK (c-Jun NH2-terminal kinase). In endothelial cells, NO increases soluble guanylate cyclase (sGC) activity, which, in turn, activates protein kinase G (PKG) by increasing cGMP levels. In this study, we investigated whether BK acts via the sGC-cGMP-PKG pathway to inhibit the negative effects of JNK on insulin signaling and glucose uptake in rat adipocytes. BK augmented cGMP concentrations. The BK-induced enhancement of insulin-stimulated glucose uptake was mimicked by the sGC activator YC-1 and a cell-permeable cGMP analog, CPT-cGMP, and inhibited by the sGC inhibitor ODQ and the PKG inhibitor KT 5823. Transfection of dominant-negative PKG reduced the BK augmentation of insulin-induced Akt phosphorylation. The activation of JNK and ERK1/2 by insulin was attenuated by BK, which was mediated by the sGC-cGMP-PKG pathway. Whereas insulin-stimulated phosphorylation of upstream activators of JNK and ERK, i.e., MKK4 and MEK1/2, was unaffected, BK augmented insulin-mediated induction of MKP-5 mRNA and protein levels. Furthermore, zaprinast, a phosphodiesterase inhibitor, enhanced cGMP and MKP-5 and prolonged the action of BK. These data indicate that BK enhances insulin action by inhibition of negative feedback by JNK and ERK via upregulation of MKP-5, mediated by the sGC-cGMP-PKG signaling pathway.
Collapse
Affiliation(s)
- María E Frigolet
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
| | - Garry Thomas
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kristin Beard
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Huogen Lu
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lijiang Liu
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - I George Fantus
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada;
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Jankovic A, Korac A, Buzadzic B, Stancic A, Otasevic V, Ferdinandy P, Daiber A, Korac B. Targeting the NO/superoxide ratio in adipose tissue: relevance to obesity and diabetes management. Br J Pharmacol 2017; 174:1570-1590. [PMID: 27079449 PMCID: PMC5446578 DOI: 10.1111/bph.13498] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/21/2022] Open
Abstract
Insulin sensitivity and metabolic homeostasis depend on the capacity of adipose tissue to take up and utilize excess glucose and fatty acids. The key aspects that determine the fuel-buffering capacity of adipose tissue depend on the physiological levels of the small redox molecule, nitric oxide (NO). In addition to impairment of NO synthesis, excessive formation of the superoxide anion (О2•- ) in adipose tissue may be an important interfering factor diverting the signalling of NO and other reactive oxygen and nitrogen species in obesity, resulting in metabolic dysfunction of adipose tissue over time. Besides its role in relief from superoxide burst, enhanced NO signalling may be responsible for the therapeutic benefits of different superoxide dismutase mimetics, in obesity and experimental diabetes models. This review summarizes the role of NO in adipose tissue and highlights the effects of NO/О2•- ratio 'teetering' as a promising pharmacological target in the metabolic syndrome. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Aleksandra Korac
- Faculty of Biology, Center for Electron MicroscopyUniversity of BelgradeBelgradeSerbia
| | - Biljana Buzadzic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Ana Stancic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Vesna Otasevic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Péter Ferdinandy
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- Pharmahungary GroupSzegedHungary
| | - Andreas Daiber
- Center for Cardiology ‐ Cardiology 1, Molecular CardiologyUniversity Medical CenterMainzGermany
| | - Bato Korac
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| |
Collapse
|
31
|
Supakankul P, Kumchoo T, Mekchay S. Identification and characterization of novel single nucleotide polymorphism markers for fat deposition in muscle tissue of pigs using amplified fragment length polymorphism. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2016; 30:338-346. [PMID: 27608636 PMCID: PMC5337912 DOI: 10.5713/ajas.16.0200] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/26/2016] [Accepted: 09/08/2016] [Indexed: 02/07/2023]
Abstract
Objective This study was conducted to identify and evaluate the effective single nucleotide polymorphism (SNP) markers for fat deposition in the longissimus dorsi muscles of pigs using the amplified fragment length polymorphism (AFLP) approach. Methods Sixty-four selective primer combinations were used to identify the AFLP markers in the 20 highest- and 20 lowest-intramuscular fat (IMF) content phenotypes. Five AFLP fragments were converted into simple codominant SNP markers. These SNP markers were tested in terms of their association with IMF content and fatty acid (FA) composition traits in 620 commercially crossbred pigs. Results The SSC7 g.4937240C>G marker showed an association with IMF content (p<0.05). The SSC9 g.5496647_5496662insdel marker showed a significant association with IMF content and arachidonic levels (p<0.05). The SSC10 g.71225134G>A marker revealed an association with palmitoleic and ω9 FA levels (p<0.05), while the SSC17 g.61976696G>T marker showed a significant association with IMF content and FA levels of palmitoleic, eicosenoic, arachidonic, monounsaturated fatty acids, and ω9 FA levels. However, no significant association of SSC8 g.47338181G>A was observed with any IMF and FA levels in this study. Conclusion Four SNP markers (SSC7 g.4937240C>G, SSC9 g.5496647_5496662insdel, SSC10 g.71225134G>A, and SSC17 g.61976696G>T) were found to be associated with IMF and/or FA content traits in commercially crossbred pigs. These findings provide evidence of the novel SNP markers as being potentially useful for selecting pigs with the desirable IMF content and FA composition.
Collapse
Affiliation(s)
- Pantaporn Supakankul
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand.,Center of Excellence on Agricultural Biotechnology: (AG-BIO/PERDO-CHE), Bangkok 10900, Thailand.,Human and Animal Biotechnology Program, Graduate School, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Tanavadee Kumchoo
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Supamit Mekchay
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
32
|
Wang X, Hai C. Redox modulation of adipocyte differentiation: hypothesis of "Redox Chain" and novel insights into intervention of adipogenesis and obesity. Free Radic Biol Med 2015; 89:99-125. [PMID: 26187871 DOI: 10.1016/j.freeradbiomed.2015.07.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 06/19/2015] [Accepted: 07/08/2015] [Indexed: 02/08/2023]
Abstract
In view of the global prevalence of obesity and obesity-associated disorders, it is important to clearly understand how adipose tissue forms. Accumulating data from various laboratories implicate that redox status is closely associated with energy metabolism. Thus, biochemical regulation of the redox system may be an attractive alternative for the treatment of obesity-related disorders. In this work, we will review the current data detailing the role of the redox system in adipocyte differentiation, as well as identifying areas for further research. The redox system affects adipogenic differentiation in an extensive way. We propose that there is a complex and interactive "redox chain," consisting of a "ROS-generating enzyme chain," "combined antioxidant chain," and "transcription factor chain," which contributes to fine-tune the regulation of ROS level and subsequent biological consequences. The roles of the redox system in adipocyte differentiation are paradoxical. The redox system exerts a "tridimensional" mechanism in the regulation of adipocyte differentiation, including transcriptional, epigenetic, and posttranslational modulations. We suggest that redoxomic techniques should be extensively applied to understand the biological effects of redox alterations in a more integrated way. A stable and standardized "redox index" is urgently needed for the evaluation of the general redox status. Therefore, more effort should be made to establish and maintain a general redox balance rather than to conduct simple prooxidant or antioxidant interventions, which have comprehensive implications.
Collapse
Affiliation(s)
- Xin Wang
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| | - Chunxu Hai
- Department of Toxicology, Shaanxi Key Lab of Free Radical Biology and Medicine, the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
33
|
Minakuchi H, Wakino S, Hosoya K, Sueyasu K, Hasegawa K, Shinozuka K, Yoshifuji A, Futatsugi K, Komatsu M, Kanda T, Tokuyama H, Hayashi K, Itoh H. The role of adipose tissue asymmetric dimethylarginine/dimethylarginine dimethylaminohydrolase pathway in adipose tissue phenotype and metabolic abnormalities in subtotally nephrectomized rats. Nephrol Dial Transplant 2015; 31:413-23. [DOI: 10.1093/ndt/gfv367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/09/2015] [Indexed: 01/12/2023] Open
|
34
|
Serum nitric oxide metabolites are associated with the risk of hypertriglyceridemic-waist phenotype in women: Tehran Lipid and Glucose Study. Nitric Oxide 2015; 50:52-57. [PMID: 26284308 DOI: 10.1016/j.niox.2015.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/12/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM There are some controversial issues regarding the association of nitric oxide and obesity-related states. This study was conducted to investigate whether serum nitric oxide metabolites (NOx) could predict the occurrence of visceral lipid accumulation, defined as hypertriglyceridemic-waist (HTW) phenotype. METHODS We used a prospective approach for this study conducted on participants of the Tehran Lipid and Glucose Study, 2243 adult men and women were followed for a median of 6.3 years. Serum NOx concentrations were measured at baseline (2006-2008), and demographics, anthropometrics and biochemical variables were evaluated at baseline and again after a 3-year (2009-2011) and a 6-year follow-up (2012-2014). The occurrence of HTW phenotype, defined as waist circumference ≥90 cm in men and ≥85 cm in women, along with serum triglyceride levels ≥177 mg/dL, were assessed across serum NOx tertiles. RESULTS Mean age of participants was 41.5 ± 14.5 years at baseline and 39.4% were male. The cumulative incidence of HTW phenotype was 37.6% (33.2% in men, 40.5% in women). There was no significant association between serum NOx and the occurrence of HTW phenotype in men. After adjustment of confounding variables, risk of HTW phenotype in women, in the highest compared to the lowest tertile of serum NOx (≥30.9 vs. <19.9 μmol/L), increased by 39% (OR = 1.39, 95% CI = 1.05-1.93, P for trend = 0.053). CONCLUSION Serum NOx level was an independent predictor of HTW phenotype in women.
Collapse
|
35
|
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
|
36
|
Johlfs MG, Gorjala P, Urasaki Y, Le TT, Fiscus RR. Capillary Isoelectric Focusing Immunoassay for Fat Cell Differentiation Proteomics. PLoS One 2015; 10:e0132105. [PMID: 26132171 PMCID: PMC4489199 DOI: 10.1371/journal.pone.0132105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/10/2015] [Indexed: 01/09/2023] Open
Abstract
Profiling cellular proteome is critical to understanding signal integration during cell fate determination. In this study, the capability of capillary isoelectric focusing (cIEF) immunoassays to detect post-translational modifications (PTM) of protein isoforms is demonstrated. cIEF immunoassays exhibit protein detection sensitivity at up to 5 orders of magnitude higher than traditional methods. This detection ultra-sensitivity permits proteomic profiling of several nanograms of tissue samples. cIEF immunoassays are employed to simultaneously profile three protein kinases during fat cell differentiation: cGMP-dependent protein kinase type I (PKG-I) of the nitric oxide (NO) signaling pathway, protein kinase B (Akt) of the insulin signaling pathway, and extracellular signal-regulated kinase (ERK) of the mitogen-activated protein kinase (MAPK) signaling pathway. Interestingly, a switch in the expression level of PKG- isoforms is observed during fat cell differentiation. While both PKG-Iα and PKG-Iβ isoforms are present in preadipocytes, only PKG-Iβ isoform is expressed in adipocytes. On the other hand, the phosphorylation level increases for Akt while decreases for ERK1 and ERK2 following the maturation of preadipocytes into adipocytes. Taken together, cIEF immunoassay provides a highly sensitive means to study fat cell differentiation proteomics. cIEF immunoassay should be a powerful proteomics tool to study complex protein signal integration in biological systems.
Collapse
Affiliation(s)
- Mary G. Johlfs
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
| | - Priyatham Gorjala
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
| | - Yasuyo Urasaki
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
| | - Thuc T. Le
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
- * E-mail: (TTL); (RRF)
| | - Ronald R. Fiscus
- Department of Biomedical Sciences, Center for Diabetes and Obesity Research, College of Medicine, Roseman University of Health Sciences, 10530 Discovery Drive, Las Vegas, Nevada, 89135, United States of America
- * E-mail: (TTL); (RRF)
| |
Collapse
|
37
|
Aquilano K, Lettieri Barbato D, Rosa CM. The multifaceted role of nitric oxide synthases in mitochondrial biogenesis and cell differentiation. Commun Integr Biol 2015; 8:e1017158. [PMID: 26479127 PMCID: PMC4594549 DOI: 10.1080/19420889.2015.1017158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 12/04/2014] [Indexed: 10/25/2022] Open
Abstract
Nitric oxide (NO) is physiologically synthetized by a family of enzymes called NO synthases (NOSs). NO is a pleiotropic second messenger having a fundamental role in several cellular processes including cell differentiation. Being a high reactive molecule, NO must be synthetized in close proximity to the effector/target. For this reason, the subcellular localization of NOSs is tightly regulated by different post-translation mechanisms. Recently, in murine C2C12 myoblasts, we have demonstrated that mitochondrial biogenesis, an essential event for cell differentiation, can be effective only if the site of NO production is located at nuclear level, where NO favors the CREB-dependent expression of PGC-1α gene. The increase of NO flux in nuclei is elicited by the up-regulation and redistribution of neuronal NOS (nNOS) toward nuclei. Herein we show that an upregulation of endothelial NOS (eNOS) occurs during adipocyte differentiation in 3T3-L1 cells. However, differently to differentiating myocytes, a concomitant redistribution of eNOS toward nuclei was not detected. We also observed that, upon treatment with the NO synthesis inhibitor L-NAME, mitochondrial biogenesis as well as triglyceride accumulation that normally occurs during adipogenesis were not impeded. The absence of eNOS in nuclei together with the ineffectiveness of L-NAME suggest that, at least during 3T3-L1 differentiation, NO is not fundamental for the induction of mitochondrial biogenesis and adipogenesis.
Collapse
Affiliation(s)
- Katia Aquilano
- Department of Biology; University of Rome "Tor Vergata" ; Rome, Italy ; IRCCS San Raffaele ; Rome, Italy
| | | | - Ciriolo Maria Rosa
- Department of Biology; University of Rome "Tor Vergata" ; Rome, Italy ; IRCCS San Raffaele ; Rome, Italy
| |
Collapse
|
38
|
Cao Y, Gomes SA, Rangel EB, Paulino EC, Fonseca TL, Li J, Teixeira MB, Gouveia CH, Bianco AC, Kapiloff MS, Balkan W, Hare JM. S-nitrosoglutathione reductase-dependent PPARγ denitrosylation participates in MSC-derived adipogenesis and osteogenesis. J Clin Invest 2015; 125:1679-91. [PMID: 25798618 DOI: 10.1172/jci73780] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/06/2015] [Indexed: 01/04/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) are a common precursor of both adipocytes and osteoblasts. While it is appreciated that PPARγ regulates the balance between adipogenesis and osteogenesis, the roles of additional regulators of this process remain controversial. Here, we show that MSCs isolated from mice lacking S-nitrosoglutathione reductase, a denitrosylase that regulates protein S-nitrosylation, exhibited decreased adipogenesis and increased osteoblastogenesis compared with WT MSCs. Consistent with this cellular phenotype, S-nitrosoglutathione reductase-deficient mice were smaller, with reduced fat mass and increased bone formation that was accompanied by elevated bone resorption. WT and S-nitrosoglutathione reductase-deficient MSCs exhibited equivalent PPARγ expression; however, S-nitrosylation of PPARγ was elevated in S-nitrosoglutathione reductase-deficient MSCs, diminishing binding to its downstream target fatty acid-binding protein 4 (FABP4). We further identified Cys 139 of PPARγ as an S-nitrosylation site and demonstrated that S-nitrosylation of PPARγ inhibits its transcriptional activity, suggesting a feedback regulation of PPARγ transcriptional activity by NO-mediated S-nitrosylation. Together, these results reveal that S-nitrosoglutathione reductase-dependent modification of PPARγ alters the balance between adipocyte and osteoblast differentiation and provides checkpoint regulation of the lineage bifurcation of these 2 lineages. Moreover, these findings provide pathophysiological and therapeutic insights regarding MSC participation in adipogenesis and osteogenesis.
Collapse
|
39
|
Oxidative stress and metabolic pathologies: from an adipocentric point of view. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:908539. [PMID: 25143800 PMCID: PMC4131099 DOI: 10.1155/2014/908539] [Citation(s) in RCA: 173] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/20/2014] [Accepted: 06/26/2014] [Indexed: 02/07/2023]
Abstract
Oxidative stress plays a pathological role in the development of various diseases including diabetes, atherosclerosis, or cancer. Systemic oxidative stress results from an imbalance between oxidants derivatives production and antioxidants defenses. Reactive oxygen species (ROS) are generally considered to be detrimental for health. However, evidences have been provided that they can act as second messengers in adaptative responses to stress. Obesity represents a major risk factor for deleterious associated pathologies such as type 2 diabetes, liver, and coronary heart diseases. Many evidences regarding obesity-induced oxidative stress accumulated over the past few years based on established correlations of biomarkers or end-products of free-radical-mediated oxidative stress with body mass index. The hypothesis that oxidative stress plays a significant role in the development of metabolic disorders, especially insulin-resistance state, is supported by several studies where treatments reducing ROS production reverse metabolic alterations, notably through improvement of insulin sensitivity, hyperlipidemia, or hepatic steatosis. In this review, we will develop the mechanistic links between oxidative stress generated by adipose tissue in the context of obesity and its impact on metabolic complications development. We will also attempt to discuss potential therapeutic approaches targeting obesity-associated oxidative stress in order to prevent associated-metabolic complications.
Collapse
|
40
|
Abstract
In adipocytes the hydrolysis of TAG to produce fatty acids and glycerol under fasting conditions or times of elevated energy demands is tightly regulated by neuroendocrine signals, resulting in the activation of lipolytic enzymes. Among the classic regulators of lipolysis, adrenergic stimulation and the insulin-mediated control of lipid mobilisation are the best known. Initially, hormone-sensitive lipase (HSL) was thought to be the rate-limiting enzyme of the first lipolytic step, while we now know that adipocyte TAG lipase is the key enzyme for lipolysis initiation. Pivotal, previously unsuspected components have also been identified at the protective interface of the lipid droplet surface and in the signalling pathways that control lipolysis. Perilipin, comparative gene identification-58 (CGI-58) and other proteins of the lipid droplet surface are currently known to be key regulators of the lipolytic machinery, protecting or exposing the TAG core of the droplet to lipases. The neuroendocrine control of lipolysis is prototypically exerted by catecholaminergic stimulation and insulin-induced suppression, both of which affect cyclic AMP levels and hence the protein kinase A-mediated phosphorylation of HSL and perilipin. Interestingly, in recent decades adipose tissue has been shown to secrete a large number of adipokines, which exert direct effects on lipolysis, while adipocytes reportedly express a wide range of receptors for signals involved in lipid mobilisation. Recently recognised mediators of lipolysis include some adipokines, structural membrane proteins, atrial natriuretic peptides, AMP-activated protein kinase and mitogen-activated protein kinase. Lipolysis needs to be reanalysed from the broader perspective of its specific physiological or pathological context since basal or stimulated lipolytic rates occur under diverse conditions and by different mechanisms.
Collapse
|
41
|
Bulloch JM, Daly CJ. Autonomic nerves and perivascular fat: interactive mechanisms. Pharmacol Ther 2014; 143:61-73. [PMID: 24560685 DOI: 10.1016/j.pharmthera.2014.02.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 12/31/2022]
Abstract
The evidence describing the autonomic innervation of body fat is reviewed with a particular focus on the role of the sympathetic neurotransmitters. In compiling the evidence, a strong case emerges for the interaction between autonomic nerves and perivascular adipose tissue (PVAT). Adipocytes have been shown to express receptors for neurotransmitters released from nearby sympathetic varicosities such as adrenoceptors (ARs), purinoceptors and receptors for neuropeptide Y (NPY). Noradrenaline can modulate both lipolysis (via α2- and β3-ARs) and lipogenesis (via α1- and β3-ARs). ATP can inhibit lipolysis (via P1 purinoceptors) or stimulate lipolysis (via P2y purinoceptors). NPY, which can be produced by adipocytes and sympathetic nerves, inhibits lipolysis. Thus the sympathetic triad of transmitters can influence adipocyte free fatty acid (FFA) content. Substance P (SP) released from sensory nerves has also been shown to promote lipolysis. Therefore, we propose a mechanism whereby sympathetic neurotransmission can simultaneously activate smooth muscle cells in the tunica media to cause vasoconstriction and alter FFA content and release from adjacent adipocytes in PVAT. The released FFA can influence endothelial function. Adipocytes also release a range of vasoactive substances, both relaxing and contractile factors, including adiponectin and reactive oxygen species. The action of adipokines (such as adiponectin) and reactive oxygen species (ROS) on cells of the vascular adventitia and nerves has yet to be fully elucidated. We hypothesise a strong link between PVAT and autonomic fibres and suggest that this poorly understood relationship is extremely important for normal vascular function and warrants a detailed study.
Collapse
Affiliation(s)
- Janette M Bulloch
- School of Science, University of the West of Scotland, Hamilton ML3 0JB, Scotland.
| | - Craig J Daly
- School of Life Sciences, University of Glasgow, Glasgow G128QQ, Scotland.
| |
Collapse
|
42
|
Sawamoto K, Huong TT, Sugimoto N, Mizutani Y, Sai Y, Miyamoto KI. Mechanisms of Lower Maintenance Dose of Tacrolimus in Obese Patients. Drug Metab Pharmacokinet 2014; 29:341-7. [DOI: 10.2133/dmpk.dmpk-13-rg-110] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
43
|
Liu GS, Chan EC, Higuchi M, Dusting GJ, Jiang F. Redox mechanisms in regulation of adipocyte differentiation: beyond a general stress response. Cells 2012; 1:976-93. [PMID: 24710538 PMCID: PMC3901142 DOI: 10.3390/cells1040976] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 10/26/2012] [Accepted: 10/31/2012] [Indexed: 02/07/2023] Open
Abstract
In this review, we summarize advances in our understanding of redox-sensitive mechanisms that regulate adipogenesis. Current evidence indicates that reactive oxygen species may act to promote both the initiation of adipocyte lineage commitment of precursor or stem cells, and the terminal differentiation of preadipocytes to mature adipose cells. These can involve redox regulation of pathways mediated by receptor tyrosine kinases, peroxisome proliferator-activated receptor γ (PPARγ), PPARγ coactivator 1α (PGC-1α), AMP-activated protein kinase (AMPK), and CCAAT/enhancer binding protein β (C/EBPβ). However, the precise roles of ROS in adipogenesis in vivo remain controversial. More studies are needed to delineate the roles of reactive oxygen species and redox signaling mechanisms, which could be either positive or negative, in the pathogenesis of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Guei-Sheung Liu
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Elsa C Chan
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Masayoshi Higuchi
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Gregory J Dusting
- Centre for Eye Research Australia, University of Melbourne, Victoria 3002, Australia.
| | - Fan Jiang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital, Shandong University, Jinan 250-012, Shandong, China.
| |
Collapse
|
44
|
Hemmrich K, Paul NE, Pallua N. The nitric oxide system--cure for shortcomings in adipose tissue engineering? J Tissue Eng Regen Med 2012; 6 Suppl 3:S1-3. [PMID: 22888023 DOI: 10.1002/term.1541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 03/04/2012] [Accepted: 04/18/2012] [Indexed: 11/07/2022]
Abstract
Adipose tissue engineering aims to grow fat tissue for soft tissue reconstruction after tumour resection or trauma. However, insufficient progenitor cell differentiation and poor vascularization compromise the generation of clinically applicable adipose tissue. The desired process of neo-adipogenesis seems to be difficult to mimic, even though it takes place in all of us, inevitably and rapidly, as soon as we start consuming high-caloric diets. It has previously been proposed that inflammation and its key regulator, nitric oxide (NO), may play a relevant part in neo-adipogenesis. We here discuss how a controlled activation of the nitric oxide system on various levels may represent a cure for several current shortcomings in adipose tissue engineering.
Collapse
Affiliation(s)
- Karsten Hemmrich
- Department of Plastic Surgery and Hand Surgery, Burn Centre, University Hospital of RWTH Aachen University of Technology, Aachen, Germany.
| | | | | |
Collapse
|
45
|
Imaging immune and metabolic cells of visceral adipose tissues with multimodal nonlinear optical microscopy. PLoS One 2012; 7:e38418. [PMID: 22701636 PMCID: PMC3372513 DOI: 10.1371/journal.pone.0038418] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 05/09/2012] [Indexed: 12/27/2022] Open
Abstract
Visceral adipose tissue (VAT) inflammation is recognized as a mechanism by which obesity is associated with metabolic diseases. The communication between adipose tissue macrophages (ATMs) and adipocytes is important to understanding the interaction between immunity and energy metabolism and its roles in obesity-induced diseases. Yet visualizing adipocytes and macrophages in complex tissues is challenging to standard imaging methods. Here, we describe the use of a multimodal nonlinear optical (NLO) microscope to characterize the composition of VATs of lean and obese mice including adipocytes, macrophages, and collagen fibrils in a label-free manner. We show that lipid metabolism processes such as lipid droplet formation, lipid droplet microvesiculation, and free fatty acids trafficking can be dynamically monitored in macrophages and adipocytes. With its versatility, NLO microscopy should be a powerful imaging tool to complement molecular characterization of the immunity-metabolism interface.
Collapse
|
46
|
Sotornik R, Brassard P, Martin E, Yale P, Carpentier AC, Ardilouze JL. Update on adipose tissue blood flow regulation. Am J Physiol Endocrinol Metab 2012; 302:E1157-70. [PMID: 22318953 DOI: 10.1152/ajpendo.00351.2011] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
According to Fick's principle, any metabolic or hormonal exchange through a given tissue depends on the product of the blood flow to that tissue and the arteriovenous difference. The proper function of adipose tissue relies on adequate adipose tissue blood flow (ATBF), which determines the influx and efflux of metabolites as well as regulatory endocrine signals. Adequate functioning of adipose tissue in intermediary metabolism requires finely tuned perfusion. Because metabolic and vascular processes are so tightly interconnected, any disruption in one will necessarily impact the other. Although altered ATBF is one consequence of expanding fat tissue, it may also aggravate the negative impacts of obesity on the body's metabolic milieu. This review attempts to summarize the current state of knowledge on adipose tissue vascular bed behavior under physiological conditions and the various factors that contribute to its regulation as well as the possible participation of altered ATBF in the pathophysiology of metabolic syndrome.
Collapse
Affiliation(s)
- Richard Sotornik
- Diabetes and Metabolism Research Group, Division of Endocrinology, Department of Medicine, Centre Hospitalier, Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
47
|
Dias JP, Couture R. Blockade of kinin B(1) receptor reverses plasma fatty acids composition changes and body and tissue fat gain in a rat model of insulin resistance. Diabetes Obes Metab 2012; 14:244-53. [PMID: 22023455 DOI: 10.1111/j.1463-1326.2011.01521.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AIM Kinin B(1) receptor (B(1) R) contributes to insulin resistance through a mechanism involving oxidative stress. This study examined the effect of B(1) R blockade on the changes in plasma fatty acids composition, body and tissue fat mass and adipose tissue inflammation that influence insulin resistance. METHODS Sprague-Dawley rats were fed with 10% D-glucose or tap water (Control) for 13 weeks and during the last week, rats were administered the B(1) R antagonist SSR240612 (10 mg/kg/day, gavage) or vehicle. The following parameters were assessed: plasma fatty acids (by gas chromatography), body composition (by EchoMRI), metabolic hormone levels (by radioimmunoassay), expression of B(1) R and inflammatory markers in adipose tissue (by Western blot and qRT-PCR). RESULTS Glucose feeding significantly increased plasma levels of glucose, insulin, leptin, palmitoleic acid (16:1n-7), oleic acid (18:1n-9), Δ6 and Δ9 desaturases while linoleic acid (18:2n-6), arachidonic acid (20:4n-6) and Δ5 desaturase were decreased. SSR240612 reduced plasma levels of insulin, glucose, the homeostasis model assessment index of insulin resistance, palmitoleic acid and n-7 family. Alterations of Δ5, Δ6 and Δ9 desaturases were normalized by SSR240612. The B(1) R antagonist also reversed the enhancing effect of glucose feeding on whole body and epididymal fat mass and on the expression of macrophage CD68, interleukin-1β, tumour necrosis factor-α and inducible nitric oxide synthase in retroperitoneal adipose tissue. B(1) R protein and mRNA were not detected in retroperitoneal adipose tissue. CONCLUSION Insulin resistance in glucose-fed rats is associated with low state inflammation in adipose tissue and plasma fatty acids changes which are reversed by B(1) R blockade. These beneficial effects may contribute to insulin sensitivity improvement and the prevention of obesity.
Collapse
Affiliation(s)
- J P Dias
- Department of Physiology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | | |
Collapse
|
48
|
Choi H, Myung K. Vitamin D₃ regulation of body fat, cytokines, and calpain gene expression. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2012; 92:632-637. [PMID: 25520983 DOI: 10.1002/jsfa.4622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
BACKGROUND We conducted an in vivo experiment to determine whether vitamin D₃ acts as a fat synthesizer and/or meat tenderizer in mice. At 6 weeks of age, 20 male C57BL/6 wild-type mice were randomly divided into two groups (10 mice per group) and fed a modified AIN93G diet with (vitamin D₃ diet) or without (basal diet) 10 IU 25-OH-cholecalciferol kg⁻³ for 3 weeks. RESULTS When vitamin D₃ was fed to mice for 3 weeks, body fat was significantly increased compared to mice fed a basal diet. There was, however, no difference in body weight between the two groups. Vitamin D₃ increased the gene expressions of pro-inflammatory cytokines and peroxisome proliferator-activated receptor gamma, but decreased interleukin-15 in adipose tissue through nuclear vitamin D receptor and uncoupling protein-2 signals. The muscle inducible nitrate oxide synthase content of mice fed vitamin D₃ was higher than those fed a basal diet, while muscle arginase l showed a reverse phenomenon. longissimuss dorsi muscle of vitamin D₃-fed mice showed more severe fat deposition than those fed a basal diet. Vitamin D₃ amplified muscle u- and m-calpain protein content and suppressed muscle calpastatin protein content. CONCLUSION These findings suggest that vitamin D3 can be used as a fat synthesizer and meat tenderizer in meat-producing animals.
Collapse
|
49
|
De Toni L, Strapazzon G, Gianesello L, Caretta N, Pilon C, Bruttocao A, Foresta C. Effects of type 5-phosphodiesterase inhibition on energy metabolism and mitochondrial biogenesis in human adipose tissue ex vivo. J Endocrinol Invest 2011; 34:738-41. [PMID: 22234177 DOI: 10.1007/bf03346724] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE An excess of adipose tissue (AT) in obese individuals is linked to increased cardiovascular risk and mitochondria have been shown to be defective in the muscle and AT of patients with metabolic disorders such as obesity and Type 2 diabetes. Nitric oxide (NO) generated by endothelial NO synthase (eNOS) plays a role in mitochondrial biogenesis through cyclic-GMP (cGMP). AT harbors the whole molecular signaling pathway of NO, together with type 5-phosphodiesterase (PDE- 5), the main cGMP catabolising enzyme. AIM Our aim was to evaluate the effect of the modulation of NO pathway, through PDE-5 inhibition, on energy metabolism and mitochondria biogenesis in human omental AT. METHODS AND MEASUREMENTS Cultured human omental AT was stimulated with PDE-5 inhibitor, vardenafil, at different concentration for 24 and 72 h. Analysis of the expression of both key-regulator genes of adipocyte metabolism and mitochondria-biogenesis markers was performed. RESULTS We found an increased gene expression of peroxisome proliferator-activated receptor-γ (PPAR-γ), adiponectin, and proliferator- activated receptor gamma coactivator-1 α (PGC-1α) after a 24-h stimulation with vardenafil at the lowest concentration employed compared to controls (p<0.05). After 72 h of stimulation, a significant increase of mitochondrial DNA was found compared to control samples (p<0.05). CONCLUSION Our data suggest that PDE-5 inhibition could have an impact on mitochondrial content of human AT suggesting a positive effect on energy metabolism and adding new elements in the comprehension of AT pathophysiology.
Collapse
Affiliation(s)
- L De Toni
- Department of Histology, Microbiology and Medical Biotechnologies, Section of Clinical Pathology and Centre for Human Reproduction Pathology, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Armani A, Marzolla V, Rosano GMC, Fabbri A, Caprio M. Phosphodiesterase type 5 (PDE5) in the adipocyte: a novel player in fat metabolism? Trends Endocrinol Metab 2011; 22:404-11. [PMID: 21741267 DOI: 10.1016/j.tem.2011.05.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 05/18/2011] [Accepted: 05/25/2011] [Indexed: 12/19/2022]
Abstract
Phosphodiesterase type 5 (PDE5) is expressed in many tissues (e.g. heart, lung, pancreas, penis) and plays a specific role in hydrolyzing cyclic guanosine monophosphate (cGMP). In adipocytes, cGMP regulates crucial functions by activating cGMP-dependent protein kinase (PKG). Interestingly, PDE5 was recently identified in adipose tissue, although its role remains unclear. Its inhibition, however, was recently shown to affect adipose differentiation and aromatase function. This review summarizes evidence supporting a role for the PDE5-regulated cGMP/PKG system in adipose tissue and its effects on adipocyte function. A better elucidation of the role of PDE5 in the adipocyte could reveal new therapeutic strategies for fighting obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Andrea Armani
- Center for Clinical and Basic Research, Scientific Institute for Research, Hospitalization and Health Care (IRCCS) San Raffaele Pisana, Rome, Italy
| | | | | | | | | |
Collapse
|