1
|
Chiti H, Peyrovi P, Ramazani A, Mazloomzadeh S, Parsamanesh N. Positive association of -420C > G single nucleotide polymorphism in resistin gene promoter with insulin resistance indices in diabetic type 2 patients. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
2
|
Punch E, Klein J, Diaba-Nuhoho P, Morawietz H, Garelnabi M. Effects of PCSK9 Targeting: Alleviating Oxidation, Inflammation, and Atherosclerosis. J Am Heart Assoc 2022; 11:e023328. [PMID: 35048716 PMCID: PMC9238481 DOI: 10.1161/jaha.121.023328] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Characterized as a chronic inflammatory disease of the large arteries, atherosclerosis is the primary cause of cardiovascular disease, the leading contributor of morbidity and mortality worldwide. Elevated plasma cholesterol levels and chronic inflammation within the arterial plaque are major mediators of plaque initiation, progression, and instability. In 2003, the protein PCSK9 (proprotein convertase subtilisin/kexin 9) was discovered to play a critical role in cholesterol regulation, thus becoming a key player in the mechanisms behind atherosclerotic plaque development. Emerging evidence suggests that PCSK9 could potentially have effects on atherosclerosis that are independent of cholesterol levels. The objective of this review was to discuss the role on PCSK9 in oxidation, inflammation, and atherosclerosis. This function activates proinflammatory cytokine production and affects oxidative modifications within atherosclerotic lesions, revealing its more significant role in atherosclerosis. Although a variety of evidence demonstrates that PCSK9 plays a role in atherosclerotic inflammation, the direct mechanism of involvement is still unknown, driving a gap in knowledge to such a predominant player in cardiovascular disease. Investigation of proteins structurally related to PCSK9 may interestingly be the link in unveiling the mechanistic role of this protein’s involvement in oxidation and inflammation. Importantly, the unique structure of PCSK9 bears structural homology to a one‐of‐a‐kind domain found in the metabolic protein resistin, which is responsible for many of the same inflammatory outcomes as PCSK9. Closing this gap in knowledge of PCSK9`s role in atherosclerotic oxidation and inflammation will provide fundamental information for understanding, preventing, and treating cardiovascular disease.
Collapse
Affiliation(s)
- Emily Punch
- Department of Chemistry University of Massachusetts Lowell MA
| | - Justus Klein
- Division of Vascular Endothelium and Microcirculation Department of Medicine III University Hospital and Medical Faculty Carl Gustav CarusTechnische Universität Dresden Germany
| | - Patrick Diaba-Nuhoho
- Division of Vascular Endothelium and Microcirculation Department of Medicine III University Hospital and Medical Faculty Carl Gustav CarusTechnische Universität Dresden Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation Department of Medicine III University Hospital and Medical Faculty Carl Gustav CarusTechnische Universität Dresden Germany
| | - Mahdi Garelnabi
- Biomedical and Nutritional Sciences University of Massachusetts Lowell MA
| |
Collapse
|
3
|
Impact of Bariatric Surgery on Adipose Tissue Biology. J Clin Med 2021; 10:jcm10235516. [PMID: 34884217 PMCID: PMC8658722 DOI: 10.3390/jcm10235516] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Bariatric surgery (BS) procedures are actually the most effective intervention to help subjects with severe obesity achieve significant and sustained weight loss. White adipose tissue (WAT) is increasingly recognized as the largest endocrine organ. Unhealthy WAT expansion through adipocyte hypertrophy has pleiotropic effects on adipocyte function and promotes obesity-associated metabolic complications. WAT dysfunction in obesity encompasses an altered adipokine secretome, unresolved inflammation, dysregulated autophagy, inappropriate extracellular matrix remodeling and insufficient angiogenic potential. In the last 10 years, accumulating evidence suggests that BS can improve the WAT function beyond reducing the fat depot sizes. The causal relationships between improved WAT function and the health benefits of BS merits further investigation. This review summarizes the current knowledge on the short-, medium- and long-term outcomes of BS on the WAT composition and function.
Collapse
|
4
|
Kumar D, Lee B, Puan KJ, Lee W, Luis BS, Yusof N, Andiappan AK, Del Rosario R, Poschmann J, Kumar P, DeLibero G, Singhal A, Prabhakar S, De Yun W, Poidinger M, Rötzschke O. Resistin expression in human monocytes is controlled by two linked promoter SNPs mediating NFKB p50/p50 binding and C-methylation. Sci Rep 2019; 9:15245. [PMID: 31645609 PMCID: PMC6811637 DOI: 10.1038/s41598-019-51592-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/03/2019] [Indexed: 02/06/2023] Open
Abstract
Resistin is a key cytokine associated with metabolic and inflammatory diseases. Especially in East Asian populations, the expression levels are strongly influenced by genetic polymorphisms. Mechanisms and functional implications of this genetic control are still unknown. By employing reporter assays, EMSA, inhibition studies, bisulphite sequencing, ChIP-Seq and gene-editing we show that the p50/p50 homodimer known to act as repressor for a number of pro-inflammatory genes plays a central role in the genetic regulation of resistin in monocytes along with promoter methylation. In the common RETN haplotype p50/p50 constitutively dampens the expression by binding to the promoter. In an Asian haplotype variant however this interaction is disrupted by the A allele of rs3219175. The SNP is in very close linkage to rs34861192, a CpG SNP, located 280 bp upstream which provides an allele-specific C-methylation site. rs34861192 is located in a 100 bp region found to be methylated in the common but not in the Asian haplotype, resulting in the latter having a higher basal expression, which also associates with elevated histone acetylation (H3K27ac). Genotype associations within cohort data of 200 East Asian individuals revealed significant associations between this haplotype and the plasma levels of factors such as TGF-b, S100B, sRAGE and IL-8 as well as with myeloid DC counts. Thus, the common RETN haplotype is tightly regulated by the epigenetic mechanism linked to p50/p50-binding. This control is lost in the Asian haplotype, which may have evolved to balance the antagonistic RETN effects on pathogen protection vs. metabolic and inflammatory disease induction.
Collapse
Affiliation(s)
- Dilip Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| | - Bernett Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Kia Joo Puan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Wendy Lee
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Boris San Luis
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Nurhashikin Yusof
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Ricardo Del Rosario
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA, 02142, USA
| | - Jeremie Poschmann
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore.,Centre de Recherche en Transplantation et Immunologie, Université de Nantes, Nantes, France
| | - Pavanish Kumar
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Gennaro DeLibero
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Amit Singhal
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research of Singapore (A*STAR), Singapore, Singapore
| | - Wang De Yun
- Department of Otolaryngology, National University of Singapore, Singapore, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Olaf Rötzschke
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore.
| |
Collapse
|
5
|
Resistin in Human Seminal Plasma: Relationship with Lipid Peroxidation, CAT Activity, GSH/GSSG Ratio, and Semen Parameters. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2192093. [PMID: 31772701 PMCID: PMC6854241 DOI: 10.1155/2019/2192093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/26/2019] [Accepted: 09/07/2019] [Indexed: 12/14/2022]
Abstract
Resistin is an adipokine involved in inflammation and able to induce the expression of other proinflammatory cytokines. It is known that, in human semen, resistin is correlated with inflammatory cytokines and sperm quality. The aim of this prospective study was to explore the potential relationship between resistin, lipid peroxidation (LPO), catalase (CAT) activity, and reduced and oxidized glutathione (GSH/GSSG) ratio in semen samples of infertile patients with leukocytospermia (no. 19), infertile patients with varicocele (no. 17), and fertile men (no. 17). Semen analysis was performed following the WHO guidelines, and sperm apoptosis and necrosis were evaluated with annexin V/propidium iodide assay. Seminal plasma samples were used to determine resistin levels by an immunological method, MDA concentration by a HPLC analysis with UV detection, GSH/GSSG ratio by an enzymatic method, CAT activity by a spectrophotometric method. The results showed that, in both groups of infertile patients, semen parameters were significantly reduced (P < 0.001) and sperm apoptosis and necrosis percentages were increased. Resistin levels were significantly higher in leukocytospermia and varicocele groups (P < 0.001 and P < 0.01, respectively) as well as MDA concentration (P < 0.001) compared to controls. The MDA level was also significantly increased in the leukocytospermia group versus the varicocele group (P < 0.05). The GSH/GSSG ratio was higher in fertile controls than the leukocytospermia group (P < 0.05) and the varicocele group (P < 0.001) and in the leukocytospermia group versus the varicocele group (P < 0.05). Both the leukocytospermia and varicocele groups showed increased values of CAT activities (P < 0.001) than controls. Briefly, the correlation between variables, calculated in the whole patient population, showed that resistin levels positively correlated with MDA levels, CAT activity, sperm apoptosis, and necrosis and negatively with sperm parameters and GSH/GSSG ratio. These results support an active role of resistin in an inflammatory process causing LPO, increase of CAT activity, and decrease of GSH/GSSG ratio in seminal plasma of infertile men vs. fertile controls.
Collapse
|
6
|
The Central Role of Biometals Maintains Oxidative Balance in the Context of Metabolic and Neurodegenerative Disorders. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8210734. [PMID: 28751933 PMCID: PMC5511683 DOI: 10.1155/2017/8210734] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/19/2017] [Accepted: 05/28/2017] [Indexed: 12/13/2022]
Abstract
Traditionally, oxidative stress as a biological aspect is defined as an imbalance between the free radical generation and antioxidant capacity of living systems. The intracellular imbalance of ions, disturbance in membrane dynamics, hypoxic conditions, and dysregulation of gene expression are all molecular pathogenic mechanisms closely associated with oxidative stress and underpin systemic changes in the body. These also include aspects such as chronic immune system activation, the impairment of cellular structure renewal, and alterations in the character of the endocrine secretion of diverse tissues. All of these mentioned features are crucial for the correct function of the various tissue types in the body. In the present review, we summarize current knowledge about the common roots of metabolic and neurodegenerative disorders induced by oxidative stress. We discuss these common roots with regard to the way that (1) the respective metal ions are involved in the maintenance of oxidative balance and (2) the metabolic and signaling disturbances of the most important biometals, such as Mg2+, Zn2+, Se2+, Fe2+, or Cu2+, can be considered as the central connection point between the pathogenesis of both types of disorders and oxidative stress.
Collapse
|
7
|
Ho KT, Hsia CP, Huang CN, Chang YH, Lin Y, Shiau MY. Association between resistin promoter -420C>G polymorphisms and producing ability with type 2 diabetes mellitus. AIMS ALLERGY AND IMMUNOLOGY 2017. [DOI: 10.3934/allergy.2017.4.181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
8
|
Sparks LM, Redman LM, Conley KE, Harper ME, Hodges A, Eroshkin A, Costford SR, Gabriel ME, Yi F, Shook C, Cornnell HH, Ravussin E, Smith SR. Differences in Mitochondrial Coupling Reveal a Novel Signature of Mitohormesis in Muscle of Healthy Individuals. J Clin Endocrinol Metab 2016; 101:4994-5003. [PMID: 27710240 PMCID: PMC5155692 DOI: 10.1210/jc.2016-2742] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
CONTEXT Reduced mitochondrial coupling (ATP/O2 [P/O]) is associated with sedentariness and insulin resistance. Interpreting the physiological relevance of P/O measured in vitro is challenging. OBJECTIVE To evaluate muscle mitochondrial function and associated transcriptional profiles in nonobese healthy individuals distinguished by their in vivo P/O. DESIGN Individuals from an ancillary study of Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy phase 2 were assessed at baseline. SETTING The study was performed at Pennington Biomedical Research Center. PARTICIPANTS Forty-seven (18 males, 26-50 y of age) sedentary, healthy nonobese individuals were divided into 2 groups based on their in vivo P/O. INTERVENTION None. Main Outcome(s): Body composition by dual-energy x-ray absorptiometry, in vivo mitochondrial function (P/O and maximal ATP synthetic capacity) by 31P-magnetic resonance spectroscopy and optical spectroscopy were measured. A muscle biopsy was performed to measure fiber type, transcriptional profiling (microarray), and protein expressions. RESULTS No differences in body composition, peak aerobic capacity, type I fiber content, or mitochondrial DNA copy number were observed between the 2 groups. Compared with the uncoupled group (lower P/O), the coupled group (higher P/O) had higher rates of maximal ATP synthetic capacity (maximal ATP synthetic capacity, P < .01). Transcriptomics analyses revealed higher expressions of genes involved in mitochondrial remodeling and the oxidative stress response in the coupled group. A trend for higher mitonuclear protein imbalance (P = .06) and an elevated mitochondrial unfolded protein response (heat shock protein 60 protein; P = .004) were also identified in the coupled group. CONCLUSIONS Higher muscle mitochondrial coupling is accompanied by an overall elevation in mitochondrial function, a novel transcriptional signature of oxidative stress and mitochondrial remodeling and indications of an mitochondrial unfolded protein response.
Collapse
Affiliation(s)
- Lauren M Sparks
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Leanne M Redman
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Kevin E Conley
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Mary-Ellen Harper
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Andrew Hodges
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Alexey Eroshkin
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Sheila R Costford
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Meghan E Gabriel
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Fanchao Yi
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Cherie Shook
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Heather H Cornnell
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Eric Ravussin
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| |
Collapse
|
9
|
Luo T, Snyder SM, Zhao B, Sullivan DK, Hamilton-Reeves J, Guthrie G, Ricketts ML, Shiverick KT, Shay N. Gene Expression Patterns Are Altered in Athymic Mice and Metabolic Syndrome Factors Are Reduced in C57BL/6J Mice Fed High-Fat Diets Supplemented with Soy Isoflavones. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:7492-7501. [PMID: 27653593 DOI: 10.1021/acs.jafc.6b03401] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Soy isoflavones exert beneficial health effects; however, their potential to ameliorate conditions associated with the metabolic syndrome (MetS) has not been studied in detail. In vitro and in vivo models were used to determine the effect of isoflavones on lipid metabolism, inflammation, and oxidative stress. In nude mice, consumption of Novasoy (NS) increased cholesterol and lipid metabolism gene expression, including Scd-1 (27.7-fold), Cyp4a14 (35.2-fold), and Cyp4a10 (9.5-fold), and reduced anti-inflammatory genes, including Cebpd (16.4-fold). A high-fat (HF) diet containing 0.4% (w/w) NS for 10 weeks significantly reduced percent weight gain (74.6 ± 2.5 vs 68.6 ± 3.5%) and hepatic lipid accumulation (20 ± 1.2 vs 27 ± 1.5%), compared to HF alone (p < 0.05) in C57BL/6J mice. NS also increased lipid oxidation and antioxidant gene expression while decreasing inflammatory cytokines. In vitro analysis in HepG2 cells revealed that genistein dose-dependently decreases oleic acid-induced lipid accumulation. Soy isoflavones may ameliorate symptoms associated with MetS via anti-inflammatory, antioxidant, and hypolipidemic modulation.
Collapse
Affiliation(s)
- Ting Luo
- Food Science and Technology, Oregon State University , Corvallis, Oregon 97330, United States
| | - Sarah M Snyder
- Food Science and Technology, Oregon State University , Corvallis, Oregon 97330, United States
| | - Bingxin Zhao
- Food Science and Technology, Oregon State University , Corvallis, Oregon 97330, United States
| | - Debra K Sullivan
- Dietetics and Nutrition, Kansas University Medical Center , Kansas City, Kansas 66160, United States
| | - Jill Hamilton-Reeves
- Dietetics and Nutrition, Kansas University Medical Center , Kansas City, Kansas 66160, United States
| | - Gregory Guthrie
- Baylor College of Medicine , Houston, Texas 77030, United States
| | - Marie-Louise Ricketts
- Agriculture, Nutrition and Veterinary Sciences, University of Nevada , Reno, Nevada 89557, United States
| | - Kathleen T Shiverick
- Pharmacology, University of Florida College of Medicine , Gainesville, Florida 32610, United States
| | - Neil Shay
- Food Science and Technology, Oregon State University , Corvallis, Oregon 97330, United States
| |
Collapse
|
10
|
Brøns C, Saltbæk PN, Friedrichsen M, Chen Y, Vaag A. Endocrine and metabolic diurnal rhythms in young adult men born small vs appropriate for gestational age. Eur J Endocrinol 2016; 175:29-40. [PMID: 27252486 DOI: 10.1530/eje-16-0177] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/20/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Sleep disturbances and alterations of diurnal endocrine rhythms are associated with increased risk of type 2 diabetes (T2D). We previously showed that young men born small for gestational age (SGA) and with increased risk of T2D have elevated fat and decreased glucose oxidation rates during nighttime. In this study, we investigated whether SGA men have an altered diurnal profile of hormones, substrates and inflammatory markers implicated in T2D pathophysiology compared with matched individuals born appropriate for gestational age (AGA). METHODS We collected hourly blood samples for 24 h, to measure levels of glucose, free fatty acids (FFA), triglycerides (TG), insulin, C-peptide, leptin, resistin, ghrelin, plasminogen activator inhibitor-1 (PAI-1), incretins (GLP-1 and GIP), and inflammatory markers (TNF-α and IL-6) in 13 young men born SGA and 11 young men born AGA. RESULTS Repeated measurements analyses were used to analyze the diurnal variations and differences between groups. The SGA subjects had increased 24-h glucose (P=0.03), glucagon (P=0.03) and resistin (P=0.003) levels with no difference in diurnal rhythms compared with AGA controls. We found significant diurnal variations in levels of blood glucose, plasma TG, FFA, insulin, C-peptide, GLP-1, GIP, leptin, visfatin, TNF-α, IL-6 and PAI-1. The variation in FFA levels differed between the groups during the evening. Plasma ghrelin and glucagon levels did not display diurnal variations. CONCLUSIONS Young men born SGA exhibit elevated 24-h blood glucose, and plasma glucagon and resistin levels with no major differences in diurnal rhythms of these or other key metabolic hormones, substrates or inflammatory markers implicated in the origin of adiposity and T2D.
Collapse
Affiliation(s)
- Charlotte Brøns
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark Steno Diabetes CenterGentofte, Denmark
| | - Pernille N Saltbæk
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark
| | - Martin Friedrichsen
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark Steno Diabetes CenterGentofte, Denmark
| | - Yan Chen
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark
| | - Allan Vaag
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark Steno Diabetes CenterGentofte, Denmark
| |
Collapse
|
11
|
Azab N, Abdel-Aziz T, Ahmed A, El-deen I. Correlation of serum resistin level with insulin resistance and severity of retinopathy in type 2 diabetes mellitus. JOURNAL OF SAUDI CHEMICAL SOCIETY 2016. [DOI: 10.1016/j.jscs.2012.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
12
|
Schleinitz D. Genetic Determination of Serum Levels of Diabetes-Associated Adipokines. Rev Diabet Stud 2016; 12:277-98. [PMID: 26859657 PMCID: PMC5275755 DOI: 10.1900/rds.2015.12.277] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue secretes an abundance of proteins. Some of these proteins are known as adipokines and adipose-derived hormones which have been linked with metabolic disorders, including type 2 diabetes, and even with cancer. Variance in serum adipokine concentration is often closely associated with an increase (obesity) or decrease (lipodystrophy) in fat tissue mass, and it is affected by age, gender, and localization of the adipose tissue. However, there may be genetic variants which, in consequence, influence the serum concentration of a certain adipokine, and thereby promote metabolic disturbances or, with regard to the "protective" allele, exert beneficial effects. This review focuses on the genetic determination of serum levels of the following adipokines: adiponectin, chemerin, leptin, progranulin, resistin, retinol binding protein 4, vaspin, adipsin, apelin, and omentin. The article reports on the latest findings from genome-wide association studies (GWAS) and candidate gene studies, showing variants located in/nearby the adipokine genes and other (non-receptor) genes. An extra chapter highlights adipokine-receptor variants. Epigenetic studies on adipokines are also addressed.
Collapse
Affiliation(s)
- Dorit Schleinitz
- Integrated Research and Treatment Center AdiposityDiseases, University of Leipzig, Liebigstr. 21, 04103 Leipzig, Germany
| |
Collapse
|
13
|
Harhay MO, Kizer JR, Criqui MH, Lima JAC, Tracy R, Bluemke DA, Kawut SM. Adipokines and the Right Ventricle: The MESA-RV Study. PLoS One 2015; 10:e0136818. [PMID: 26348768 PMCID: PMC4562601 DOI: 10.1371/journal.pone.0136818] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 08/10/2015] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Obesity is associated with changes in both right (RV) and left (LV) ventricular morphology, but the biological basis of this finding is not well established. We examined whether adipokine levels were associated with RV morphology and function in a population-based multiethnic sample free of clinical cardiovascular disease. METHODS We examined relationships of leptin, resistin, TNF-α, and adiponectin with RV morphology and function (from cardiac MRI) in participants (n = 1,267) free of clinical cardiovascular disease from the Multi-Ethnic Study of Atherosclerosis (MESA)-RV study. Multivariable regressions (linear, quantile [25th and 75th] and generalized additive models [GAM]) were used to examine the independent association of each adipokine with RV mass, RV end-diastolic volume (RVEDV), RV end-systolic volume (RVESV), RV stroke volume (RVSV) and RV ejection fraction (RVEF). RESULTS Higher leptin levels were associated with significantly lower levels of RV mass, RVEDV, RVESV and stroke volume, but not RVEF, after adjustment for age, gender, race, height and weight. These associations were somewhat attenuated but still significant after adjustment for traditional risk factors and covariates, and were completely attenuated when correcting for the respective LV measures. There were no significant interactions of age, gender, or race/ethnicity on the relationship between the four adipokines and RV structure or function. CONCLUSIONS Leptin levels are associated with favorable RV morphology in a multi-ethnic population free of cardiovascular disease, however these associations may be explained by a yet to be understood bi-ventricular process as this association was no longer present after adjustment for LV values. These findings complement the associations previously shown between adipokines and LV structure and function in both healthy and diseased patients. The mechanisms linking adipokines to healthy cardiovascular function require further investigation.
Collapse
Affiliation(s)
- Michael O. Harhay
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jorge R. Kizer
- Department of Medicine, Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Michael H. Criqui
- Department of Family Medicine and Public Health, University of California San Diego, San Diego, California, United States of America
| | - João A. C. Lima
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Russell Tracy
- Department of Laboratory Medicine, University of Vermont School of Medicine, Burlington, Vermont, United States of America
| | - David A. Bluemke
- Radiology and Imaging Sciences, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health/Clinical Center, Bethesda, Maryland, United States of America
| | - Steven M. Kawut
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
14
|
Lifestyle advice follow-up improve glycemic control, redox and inflammatory status in patients with type 2 diabetes. J Diabetes Metab Disord 2014; 13:122. [PMID: 25551102 PMCID: PMC4279685 DOI: 10.1186/s40200-014-0122-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 12/09/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND The dietary composition associated to physical activity could play a significant role in improving insulin sensitivity and reducing risk of diabetes and its complications. This study was designed to investigate whether glycemic control, redox and inflammatory status impairments in patients with type 2 diabetes (T2D), were improved after 90 (d90) and 180 (d180) days follow-up of nutritional advices. METHODS Patients with T2D (n = 85) aged of 50 ± 8 years (Female/Male, 45/40), treated with oral antidiabetics (OAD) alone, with a body mass index (BMI) of 26 ± 2, were recruited. At the beginning of the study (d0), patients were instructed to follow-up nutritional advices adapted to T2D, and 30 to 45 min of walking per day. Assays were realized at d90 and d180 of follow-up. Data were compared by student 't' test and Pearson's correlation coefficients were determined between biochemical parameters and nutritional advices follow-up. RESULTS Reduced glycated haemoglobin (HbA1c), glucose and total cholesterol (TC) were noted in patients with T2D, at d90 and d180 compared to d0. Thiobarbituric acid reactive substances (TBARS) and hydroperoxyde levels were lower at d90 and d180 than d0. Serum nitric oxide (NO) was decreased at d180 compared to d0 and d90. In erythrocytes, superoxide dismutase (SOD) activity increased by 7% at d180 vs d0. Moreover, activity of glutathione peroxidase (GPx) enhanced (P < 0.05), whereas that of glutathione reductase (GRed) decreased (P < 0.001) at d90 vs d0. Resistin values were lower at d180 than d0 and d90 (P < 0.001). A progressive decrease in tumor necrosis factor-α (TNF-α) was observed at d90 and d180 vs d0. CONCLUSION Nutritional advices associated to physical activity improve glycemic control, serum TC, redox and inflammatory status in T2D, in particular after 3 months of counseling. However, these results need to be supported with a longer dietary treatment and more rigorous control during the follow-up.
Collapse
|
15
|
Singh A, Suragani M, Krishna A. Effects of resistin on ovarian folliculogenesis and steroidogenesis in the vespertilionid bat, Scotophilus heathi. Gen Comp Endocrinol 2014; 208:73-84. [PMID: 25241398 DOI: 10.1016/j.ygcen.2014.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 08/01/2014] [Accepted: 09/04/2014] [Indexed: 12/15/2022]
Abstract
The bat Scotophilus heathi exhibit prolonged anovulatory condition known as delayed ovulation coinciding with the period of extensive fat accumulation. The present study was undertaken to find out whether extensive accumulation of fat in S. heathi is responsible for suppression of ovarian activity by increasing production of adipokine resistin in the bat. This was achieved by (a) investigating variation in serum resistin level in relation to the changes in the body fat mass and (b) evaluating the effect of resistin treatment on ovarian activity with reference to steroid synthesis. An attempt was also made to determine whether resistin mediate its effects on ovary through signal transducer and activator of transcription 3 (STAT3) signaling mechanism. The results showed significant seasonal variation in serum resistin level with the peak level coinciding with the period of maximum fat accumulation, high circulating androgen level and period of anovulation. The treatment with resistin to the bat caused increase in androstenedione due to stimulatory effects on 3β-hydroxysteroid dehydrogenase, but decrease in estradiol level due to inhibitory effect on aromatase. Resistin treatment increased androgen receptor protein together with increased insulin receptor but not through conventional luteinizing hormone receptor and steroidogenic acute regulatory protein mediated pathways. This study further showed that resistin treatment increases androstenedione synthesis and up-regulates insulin receptor in the ovary through STAT3 mediated pathways. These findings suggest that obese women through increased resistin synthesis may causes development of non-ovulatory antral follicles through insulin receptor signaling cascade.
Collapse
Affiliation(s)
- Ajit Singh
- Reproductive Endocrinology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Madhuri Suragani
- Dr. Reddy's Institute of Life Sciences, Hyderabad University Campus, Gachibowly, Hyderabad 500046, India
| | - Amitabh Krishna
- Reproductive Endocrinology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
16
|
Murad A, Hassan H, Husein H, Ayad A. Serum resistin levels in nonalcoholic fatty liver disease and their relationship to severity of liver disease. JOURNAL OF ENDOCRINOLOGY, METABOLISM AND DIABETES OF SOUTH AFRICA 2014. [DOI: 10.1080/22201009.2010.10872225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
17
|
Ortega L, Navarro P, Riestra P, Gavela-Pérez T, Soriano-Guillén L, Garcés C. Association of resistin polymorphisms with resistin levels and lipid profile in children. Mol Biol Rep 2014; 41:7659-64. [PMID: 25096511 DOI: 10.1007/s11033-014-3658-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 07/27/2014] [Indexed: 12/19/2022]
Abstract
Previous research has found a correlation between resistin and lipid level variations. Polymorphisms in the resistin gene (RETN) could be involved in this relationship, but the results of the different studies are contradictory. The aim of this study was to examine the association between resistin and lipid levels, and to determine whether resistin polymorphisms are associated with resistin levels and lipid profile in prepubertal children and adolescents. The single nucleotide polymorphisms (SNPs) rs1862513 and rs10401670 were analyzed in 442 randomly selected 6- to 8-year-old children and 827 children aged 12-16 years. Anthropometric data were recorded. Lipid profile was determined using standard methods. Serum resistin levels were measured using a multiplexed bead immunoassay. Resistin polymorphisms were determined by TaqMan(®) allelic discrimination assays. A relationship was found between serum levels of resistin and the SNP rs10401670 in 6- to 8-year-old boys. SNP rs10401670 was also related to TC and LDL-cholesterol in 12- to 16-year-old boys and to HDL-C in 12- to 16-year-old girls. SNP rs1862513 was not related to any of the studied variables. Serum resistin levels were significantly and negatively associated with ApoAI levels in 12- to 16-year-old girls. A SNP in the 3'UTR region of RETN (rs10401670) is associated with resistin levels and lipid profile in children, showing different associations depending on age and gender.
Collapse
Affiliation(s)
- Lorena Ortega
- Lipid Research Laboratory, IIS-Fundación Jiménez Díaz, Avda. Reyes Católicos, 2., 28040, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
BACKGROUND AND OBJECTIVES Resistin is an adipocytokine, which has been studied for its role in insulin resistance and recently in inflammation. Several single-nucleotide polymorphisms (SNPs) have been identified in the human resistin gene (RETN). This study aims to investigate the association of RETN rs1862513 (C-420G) and rs3745367 (G+299A) SNPs with the colon cancer risk in Saudi patients. DESIGN AND SETTING This is a case-control study conducted among Saudi adult colon cancer patients recruited from King Abdulaziz Hospital and Oncology Center in Jeddah, Saudi Arabia. SUBJECTS AND METHODS In this study, 120 Saudi volunteers (60 colon cancer patients and 60 disease-free controls) were studied. The SNPs were determined by polymerase chain reaction (PCR) and genotyping using PCR- restriction fragment length polymorphism analysis. RESULTS In comparing the result obtained for the patient group with that of the controls, colon cancer group displayed different genotype distribution of the RETN C-420G and G+299A SNPs. The study indicated that the SNP-420 heterozygous (CG) genotype (odds ratio [OR]=2.48, 95% CI 1.07-5.74, P=.03) and the SNP +299 heterozygous (GA) genotype (OR=6.5, 95% CI 1.77-24.18, P=.002) significantly increased the risk of colon cancer. A further analysis of the genotype combination of SNPs RETN C-420G and G+299A showed a larger increase in the colon cancer risk. CONCLUSION These preliminary results suggested a potential role for RETN C-420G and G+299A polymorphisms in the genetic predisposition to colon cancer disease.
Collapse
Affiliation(s)
- Rowyda N Alharithy
- Dr. Rowyda N. Alharithy, Department of Biochemistry, King Abdulaziz University, PO Box 40288 Jeddah 21499 Saudi Arabia, T: 0505444238 F: 0126393640,
| |
Collapse
|
19
|
Chung CM, Lin TH, Chen JW, Leu HB, Yin WH, Ho HY, Sheu SH, Tsai WC, Chen JH, Lin SJ, Pan WH. Common quantitative trait locus downstream of RETN gene identified by genome-wide association study is associated with risk of type 2 diabetes mellitus in Han Chinese: a Mendelian randomization effect. Diabetes Metab Res Rev 2014; 30:232-40. [PMID: 24123702 DOI: 10.1002/dmrr.2481] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 09/16/2013] [Accepted: 09/27/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Plasma resistin level is a potential molecular link between obesity and diabetes. Causal role of resistin, type 2 diabetes mellitus (T2DM) and genetic variants have not been thoroughly investigated. Therefore, we conducted a genome-wide association study (GWAS) to identify quantitative trait loci associated with resistin levels and investigated whether these variants were prospectively associated with the development of metabolic syndrome (MetS) and T2DM in an independent community-based cohort, the CardioVascular Disease risk FACtors Two-township Study (CVDFACTS). RESEARCH DESIGN AND METHODS We genotyped 382 young-onset hypertensive (YOH) subjects with Illumina HumanHap550 chips and searched for quantitative trait loci (QTLs) of resistin in the 1(st) stage GWAS and confirmed the finding in another 559 YOH subjects. Logistic regression was used to examine the Mendelian randomization effects between genotypes of confirmed QTLs and metabolic outcomes in 3400 subjects of CVDFACTS. RESULTS Two single nucleotide polymorphisms (SNP) (rs3745367 and rs1423096) were significantly associated with resistin levels (p = 5.52 × 10(-15) and p = 2.54 × 10(-20) ) and replicated in another 559 YOH subjects (p = 1.29 × 10(-3) and p = 1.13 × 10(-7) ), respectively. The SNP rs1423096 was further associated with the levels of HDL-C (p = 0.006), the risk of MetS (OR = 2.21, p = 0.0034) and T2DM (OR = 1.62, p = 0.0063) in the CVDFACTS. People with the haplotypes A-G and G-G determined by rs3745367 and rs1423096 showed a significantly increased T2DM risk (p = 0.0068 and p = 0.0035, respectively) compared with those with A-A haplotype. CONCLUSION We have found that rs3745367 and rs1423096 on the RETN gene were significantly associated with resistin levels. However, rs1423096, downstream of RETN, seems to be associated with MetS and T2DM risk more so than rs3745367. The established genotype-disease association points to a causal association of resistin and T2DM.
Collapse
Affiliation(s)
- Chia-Min Chung
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Division of Health Service Research and Preventive Medicine, Institute of Population Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Acute intake of a high-fructose diet alters the balance of adipokine concentrations and induces neutrophil influx in the liver. J Nutr Biochem 2013; 25:388-94. [PMID: 24485988 DOI: 10.1016/j.jnutbio.2013.11.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 11/22/2013] [Accepted: 11/25/2013] [Indexed: 12/28/2022]
Abstract
The postprandial state is a period of metabolic fluxes, biosynthesis and oxidative metabolism. A considerable amount is known about the inflammatory response to the chronic consumption of fructose, but little is known about its effects in the postprandial state. The aim of the present study was to investigate the inflammatory effects of a single meal containing fructose on healthy mice. Male BALB/c and LysM-eGFP mice at 12-14 weeks were divided into three groups: fasted, control (mice fed with a sucrose-containing diet) and fructose (mice fed with a fructose-containing diet). One, 2 or 4 h postprandial, the BALB/c mice were killed, and samples were collected. LysM-eGFP mice were submitted to intravital microscopy. The fed mice showed a low-grade inflammatory response apart from dietary composition, which was characterized by increased numbers of leukocytes and high serum concentrations of pentraxin 3, leptin and resistin. TNF-α and CCL2 concentrations rose in the liver after the meal. IL-6 concentration increased and IL-10 decreased in the adipose tissue of the fed mice. Mice fed with the fructose-containing diet showed an intensification of the inflammatory response. Furthermore, the adiponectin concentration dropped, and the liver influx of neutrophils increased after fructose intake. Overall, this study showed a rapid increase in the systemic and tissue-specific immune response after a balanced meal. The study also showed an increased neutrophil influx in liver associated with an imbalance of adipokine concentrations and an increase of cytokine in the liver and adipose tissue following a fructose-containing meal.
Collapse
|
21
|
Roberts CK, Hevener AL, Barnard RJ. Metabolic syndrome and insulin resistance: underlying causes and modification by exercise training. Compr Physiol 2013; 3:1-58. [PMID: 23720280 DOI: 10.1002/cphy.c110062] [Citation(s) in RCA: 304] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metabolic syndrome (MS) is a collection of cardiometabolic risk factors that includes obesity, insulin resistance, hypertension, and dyslipidemia. Although there has been significant debate regarding the criteria and concept of the syndrome, this clustering of risk factors is unequivocally linked to an increased risk of developing type 2 diabetes and cardiovascular disease. Regardless of the true definition, based on current population estimates, nearly 100 million have MS. It is often characterized by insulin resistance, which some have suggested is a major underpinning link between physical inactivity and MS. The purpose of this review is to: (i) provide an overview of the history, causes and clinical aspects of MS, (ii) review the molecular mechanisms of insulin action and the causes of insulin resistance, and (iii) discuss the epidemiological and intervention data on the effects of exercise on MS and insulin sensitivity.
Collapse
Affiliation(s)
- Christian K Roberts
- Exercise and Metabolic Disease Research Laboratory, Translational Sciences Section, School of Nursing, University of California at Los Angeles, Los Angeles, California, USA.
| | | | | |
Collapse
|
22
|
Aknc A, Karakurt C, Gurbuz S, Elkran O, Nalbantoglu O, Kocak G, Guldur T, Yologlu S. Association of cardiac changes with serum adiponectin and resistin levels in obese and overweight children. J Cardiovasc Med (Hagerstown) 2013; 14:228-34. [PMID: 22441215 DOI: 10.2459/jcm.0b013e328351674e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES To investigate serum adiponectin and resistin levels in childhood obesity and their relationship with cardiac changes and insulin resistance. METHODS Seventy-one obese and 24 overweight children and 40 healthy children and adolescents were selected for the study. Height and weight measurements, BMI values and BMI SD score values were obtained for each individual. After blood pressure measurement, left ventricular wall thickness, left ventricular mass, stroke volume, cardiac output, systolic and diastolic functions of the left ventricle were measured using an M-mode, two dimensional color-coded echocardiography device. Blood samples of the individuals were obtained for fasting blood sugar, total blood cholesterol, triglyceride, low-density lipoprotein (LDL) cholesterol, very low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, insulin, C-peptide, adiponectin and resistin values. RESULTS Cholesterol and LDL values, homeostasis model assessment of insulin resistance, fasting insulin and fasting C-peptide values of the obese and overweight groups were higher (P<0.01). Adiponectin level (P<0.001) and resistin level (P<0.05) of the obese and overweight groups were lower than those of the control group (P<0.05). Echocardiographic evaluation showed diastolic dysfunction in addition to increased left ventricular wall thickness and left ventricle mass values in the obese and overweight children. We also detected a significant positive correlation among left ventricular mass, interventricular septum systolic diameter and resistin in obese children. Among the factors, resistin level was determined as an independent predictor of left ventricular mass in obese children. CONCLUSION In this study, even in asymptomatic obese and overweight children, cardiac structural and functional changes, such as increased left ventricular mass and diastolic dysfunction, were demonstrated. Although decreased adiponectin level was not related to cardiac changes, it was shown that decreased serum resistin levels in the obese cases lead to left ventricle hypertrophy.
Collapse
Affiliation(s)
- Ayşehan Aknc
- Department of Pediatric Endocrinology and Metabolism, Inonu University Medical School, Malatya, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
AL-Suhaimi EA, Shehzad A. Leptin, resistin and visfatin: the missing link between endocrine metabolic disorders and immunity. Eur J Med Res 2013; 18:12. [PMID: 23634778 PMCID: PMC3655867 DOI: 10.1186/2047-783x-18-12] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Accepted: 04/03/2013] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue is still regarded as a principle site for lipid storage and mobilizing tissue with an important role in the control of energy homeostasis. Additionally, adipose tissue-secreted hormones such as leptin, visfatin, resistin, apelin, omentin, sex steroids, and various growth factors are now regarded as a functional part of the endocrine system. These hormones also play an important role in the immune system. Several in vitro and in vivo studies have suggested the complex role of adipocyte-derived hormones in immune system and inflammation. Adipokines mediate beneficial and detrimental effects in immunity and inflammation. Many of these adipocytokines have a physiological role in metabolism. The uncontrolled secretions of several adipocytokines were associated with the stimulation of inflammatory processes leading to metabolic disorders including obesity, atherosclerosis, insulin resistance and type 2 diabetes. Obesity leads to the dysfunction of adipocytes andcorrelated with the imbalance of adipokines levels. In obese and diabetic conditions, leptin deficiency inhibited the Jak/Stat3/PI3K and insulin pathways. In this review, ample evidence exists to support the recognition of the adipocyte's role in various tissues and pathologies. New integral insights may add dimensions to translate any potential agents into the future clinical armamentarium of chronic endocrine metabolic and inflammatory diseases. Functional balance of both adipocytes and immune cells is important to exert their effects on endocrine metabolic disorders; furthermore, adipose tissue should be renamed not only as a functional part of the endocrine system but also as a new part of the immune system.
Collapse
Affiliation(s)
- Ebtesam A AL-Suhaimi
- Department of Biology, Sciences College, University of Dammam, Dammam, Saudi Arabia
| | - Adeeb Shehzad
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 702-701, South Korea
| |
Collapse
|
24
|
Makni E, Moalla W, Benezzeddine-Boussaidi L, Lac G, Tabka Z, Elloumi M. Correlation of resistin with inflammatory and cardiometabolic markers in obese adolescents with and without metabolic syndrome. Obes Facts 2013; 6:393-404. [PMID: 23970148 PMCID: PMC5644748 DOI: 10.1159/000354574] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Accepted: 10/25/2012] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The link between plasma resistin and obesity-related cardiometabolic disorders in children remains debatable. This study assessed the relationships of plasma resistin with cardiovascular risk factors, pro-inflammatory markers and insulin resistance index (HOMA-IR) in obese (Ob) adolescents and obese adolescents with metabolic syndrome (Ob-MS) compared to healthy controls (CO). METHODS 114 obese adolescents (60 Ob, age 13.6 ± 0.9 years, BMI 28.0 ± 2.2 kg/m(2), and 54 Ob-MS, age 13.8 ± 1.0 years, BMI 32.5 ± 4.8 kg/m(2)) and 37 CO (age 13.7 ± 0.8 years, BMI 22.8 ± 0.8 kg/m(2)) were studied. Anthropometrics, cardiac variables as well as fasting plasma concentrations of lipids, glucose, insulin, and adipocytokines (resistin, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), C-reactive protein (CRP)) were measured. HOMA-IR was calculated, and the presence of MS was assessed. RESULTS Plasma resistin was significantly higher in Ob-MS than in both Ob and CO and was correlated with anthropometric, cardiovascular, pro-inflammatory markers and several components of MS as was HOMA-IR in Ob and Ob-MS. With increasing the number of MS components, plasma resistin, pro-inflammatory markers, and HOMA-IR were also increased. Multiple regression models highlighted significant correlation between resistin and both HOMA-IR (r = 0.40, p < 0.05) and systolic blood pressure (r = 0.63, p < 0.01) in Ob-MS. CONCLUSION These results support the hypothesis that there is an association between circulating resistin and childhood obesity-related inflammatory and cardiometabolic events.
Collapse
Affiliation(s)
- Emna Makni
- Laboratory of Cardiocirculatory, Respiratory, and Hormonal Adaptations to Muscular Exercise, Faculty of Medicine Ibn El Jazzar, University of Sousse, Sousse, Tunisia
| | - Wassim Moalla
- Laboratory of Cardiocirculatory, Respiratory, and Hormonal Adaptations to Muscular Exercise, Faculty of Medicine Ibn El Jazzar, University of Sousse, Sousse, Tunisia
- UR EM2S, ISSEP Sfax, Tunisia
| | - Lamia Benezzeddine-Boussaidi
- Laboratory of Cardiocirculatory, Respiratory, and Hormonal Adaptations to Muscular Exercise, Faculty of Medicine Ibn El Jazzar, University of Sousse, Sousse, Tunisia
| | - Gérard Lac
- Laboratory AME2P, EA 3533, BP 10448, Clermont University Blaise Pascal, Clermont-Ferrand, France
| | - Zouhair Tabka
- Laboratory of Cardiocirculatory, Respiratory, and Hormonal Adaptations to Muscular Exercise, Faculty of Medicine Ibn El Jazzar, University of Sousse, Sousse, Tunisia
| | - Mohammed Elloumi
- Laboratory of Cardiocirculatory, Respiratory, and Hormonal Adaptations to Muscular Exercise, Faculty of Medicine Ibn El Jazzar, University of Sousse, Sousse, Tunisia
- Laboratory AME2P, EA 3533, BP 10448, Clermont University Blaise Pascal, Clermont-Ferrand, France
- *Mohamed Elloumi, Laboratory of Cardiocirculatory, Respiratory, and Hormonal Adaptations to, Muscular Exercise, Faculty of Medicine Ibn El Jazzar, University of Sousse, 4000 Sousse (Tunisia),
| |
Collapse
|
25
|
Beckers S, Zegers D, Van Camp JK, Boudin E, Nielsen TL, Brixen K, Andersen M, Van Hul W. Resistin polymorphisms show associations with obesity, but not with bone parameters in men: results from the Odense Androgen Study. Mol Biol Rep 2012. [PMID: 23203410 DOI: 10.1007/s11033-012-2327-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Resistin is an obesity-related adipokine which has also been implicated in bone metabolism. Therefore, we designed a study to investigate the possible role of resistin gene variation in both obesity and bone mineral density. We included 1,155 individuals from the Odense Androgen Study (663 young subjects and 492 older subjects), a population-based, prospective, observational study on the inter-relationship between endocrine status, body composition, muscle function, and bone metabolism in men, in an association study with resistin (RETN) polymorphisms. Three RETN variants (rs1862513, rs3745367 and rs3745369) were genotyped with TaqMan Pre-Designed Genotyping assays. Linear regression was performed to investigate the possible association of these variants with several obesity- and bone-related parameters. After genotyping 1,155 Danish men, 663 young subjects and 492 older subjects, we found that rs3745367 was associated with several obesity-related measures in both the young and elderly cohort. Rs3745369 was only associated with obesity-phenotypes in the elderly cohort. When studying the combined cohorts, we could confirm the associations of rs3745367 with several obesity-related parameters. We were unable to identify any association between RETN polymorphisms and bone-related measurements. Together, these results illustrate resistin's role in the development of obesity. Rs3745367 gives the most consistent results in the current study and these should be confirmed in other populations. Research into its possible functional effect might also be required. A role for RETN variants in determining bone mineral density seems unlikely from our results.
Collapse
Affiliation(s)
- Sigri Beckers
- Centre for Medical Genetics, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
El-Shal AS, Pasha HF, Rashad NM. Association of resistin gene polymorphisms with insulin resistance in Egyptian obese patients. Gene 2012. [PMID: 23178185 DOI: 10.1016/j.gene.2012.09.136] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Obesity associated insulin resistance is a major risk factor for type 2 diabetes mellitus. Resistin is recently reported to provide a link between obesity, insulin resistance and type 2 diabetes mellitus. We aimed to investigate the possible associations of resistin gene (RETN) polymorphisms with obesity, and to detect whether these polymorphisms are associated with glucose intolerance and type 2 diabetes mellitus in obese patients. METHODS One hundred and forty-five Egyptian obese patients with or without glucose intolerance and 155 unrelated healthy controls were enrolled in this study. Polymorphisms of RETN +299G>A and RETN -420 C>G gene were detected by polymerase chain reaction - restriction fragment length polymorphism (PCR-RFLP). Serum resistin was measured by ELISA. RESULTS RETN +299 AA and RETN -420 GG genotypes were significantly associated with obesity in Egyptian population. Moreover, the mutant alleles or genotypes of both examined polymorphisms were associated with impaired glucose tolerance and diabetes mellitus compared to normal glucose tolerant obese patients. Furthermore, our results revealed elevated waist/hip ratio, BMI, blood pressure, fasting blood glucose level, HOMA-IR, triglycerides, total cholesterol, resistin level, and decreased HDL cholesterol level in homozygote mutant genotypes carriers of both RETN polymorphisms among obese patients. CONCLUSION Resistin gene polymorphisms may play an important role in pathogenesis and susceptibility to obesity, impaired glucose tolerance, and type 2 diabetes mellitus in Egyptian population.
Collapse
Affiliation(s)
- Amal S El-Shal
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | | | | |
Collapse
|
27
|
Inflammatory effects of resistin on human smooth muscle cells: up-regulation of fractalkine and its receptor, CX3CR1 expression by TLR4 and Gi-protein pathways. Cell Tissue Res 2012; 351:161-74. [PMID: 23086480 DOI: 10.1007/s00441-012-1510-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/20/2012] [Indexed: 10/27/2022]
Abstract
In the atherosclerotic plaque, smooth muscle cells (SMC) acquire an inflammatory phenotype. Resistin and fractalkine (CX3CL1) are found in human atheroma and not in normal arteries. CX3CL1 and CX3CR1 are predominately associated with SMC. We have questioned whether resistin has a role in the expression of CX3CL1 and CX3CR1 in SMC thus contributing to the pro-inflammatory status of these cells. Cultured human aortic SMC were stimulated with 100 ng/ml resistin for 4, 6, 12, and 24 h, and then CX3CL1 and CX3CR1 expression was assessed by quantitative reverse transcription with the polymerase chain reaction and Western blot. We found that resistin up-regulated CX3CL1 and CX3CR1 in SMC and induced the phosphorylation of p38MAPK and STAT3. Inhibitors of p38MAPK, JAK-STAT, NF-kB, and AP-1 significantly reduced CX3CL1 and CX3CR1 expression. Knockdown of STAT1 and STAT3 with decoy oligodeoxinucleotides and the silencing of p65 and cjun with short interfering RNA decreased CX3CL1 and CX3CR1 expression. Anti-TLR4 antibody and pertussis toxin also reduced CX3CL1 and CX3CR1 protein expression. xCELLigence experiments revealed that resistin probably uses Gi-proteins for its effect on SMC. The CX3CL1 induced by resistin exhibited a chemotactic effect on monocyte transmigration. Thus, (1) resistin contributes to the pro-inflammatory state of SMC by the up-regulation of CX3CL1 and CX3CR1 expression via a mechanism involving NF-kB, AP-1, and STAT1/3 transcription factors, (2) resistin employs TLR4 and Gi-protein signaling for its effect on SMC, (3) CX3CL1 induced by resistin is functional in monocyte chemotaxis. The data reveal new mechanisms by which resistin promotes the inflammatory phenotype of SMC.
Collapse
|
28
|
Boumaiza I, Omezzine A, Rejeb J, Rebhi L, Ben Rejeb N, Nabli N, Ben Abdelaziz A, Bouslama A. Association between four resistin polymorphisms, obesity, and metabolic syndrome parameters in Tunisian volunteers. Genet Test Mol Biomarkers 2012; 16:1356-62. [PMID: 23020084 DOI: 10.1089/gtmb.2012.0156] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Resistin is an adipocyte-secreted cytokine recently discovered and has been proposed as a link between obesity and diabetes. Many resistin gene polymorphisms were described and their implication in obesity and metabolic syndrome (MetS) was controversial. Our aim was to study the relationship between four resistin polymorphisms (420C/G, 44G/A, 62G/A, and 394C/G), MetS parameters, and the risk of obesity in Tunisian volunteers. We recruited 169 nonobese (sex ratio=0.594; mean age=43.25±13.12 years; mean body mass index [BMI]=24.73±3.50 kg/m(2)) and 160 obese subjects (sex ratio=0.221; mean age=48.41±10.92 years; mean BMI=36.6±4.8 kg/m(2)). Genotyping was performed using polymerase chain reaction-restriction fragment length polymorphism. Anthropometric parameters, lipid levels, glycemia, and insulinemia were measured. BMI was calculated and insulin resistance was evaluated with the homeostasis model assessment insulin resistance (HOMA-IR). Statistical analyses were performed by SPSS 17.0. The 420C/G seems to contribute to obesity. In fact adjusted odds ratio (OR) of obesity associated to mutated genotypes was 2.17 and 95% confidence interval was 1.28-3.68 (p=0.004). Mutated genotypes at 420C/G were associated with higher waist circumference and BMI. In addition, 44G/A polymorphism was associated with increased total cholesterol and low-density lipoprotein-cholesterol levels. The other genotypes showed no association with MetS parameters. Concerning association between single-nucleotide polymorphisms and MetS risk, only mutated genotypes at 44G/A increase the risk of MetS after adjustment to confounding parameters (OR=1.93, p=0.023). In conclusion, resistin gene polymorphisms 420C/G and 44G/A were associated with obesity and MetS parameters in Tunisian volunteers.
Collapse
Affiliation(s)
- Imen Boumaiza
- Biochemistry Department, UR MSP 28/04, Sahloul University Hospital, Sousse, Tunisia
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Snyder EE, Walts B, Pérusse L, Chagnon YC, Weisnagel SJ, Rankinen T, Bouchard C. The Human Obesity Gene Map: The 2003 Update. ACTA ACUST UNITED AC 2012; 12:369-439. [PMID: 15044658 DOI: 10.1038/oby.2004.47] [Citation(s) in RCA: 207] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
This is the tenth update of the human obesity gene map, incorporating published results up to the end of October 2003 and continuing the previous format. Evidence from single-gene mutation obesity cases, Mendelian disorders exhibiting obesity as a clinical feature, quantitative trait loci (QTLs) from human genome-wide scans and animal crossbreeding experiments, and association and linkage studies with candidate genes and other markers is reviewed. Transgenic and knockout murine models relevant to obesity are also incorporated (N = 55). As of October 2003, 41 Mendelian syndromes relevant to human obesity have been mapped to a genomic region, and causal genes or strong candidates have been identified for most of these syndromes. QTLs reported from animal models currently number 183. There are 208 human QTLs for obesity phenotypes from genome-wide scans and candidate regions in targeted studies. A total of 35 genomic regions harbor QTLs replicated among two to five studies. Attempts to relate DNA sequence variation in specific genes to obesity phenotypes continue to grow, with 272 studies reporting positive associations with 90 candidate genes. Fifteen such candidate genes are supported by at least five positive studies. The obesity gene map shows putative loci on all chromosomes except Y. Overall, more than 430 genes, markers, and chromosomal regions have been associated or linked with human obesity phenotypes. The electronic version of the map with links to useful sites can be found at http://obesitygene.pbrc.edu.
Collapse
Affiliation(s)
- Eric E Snyder
- Human Genomics Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana 70808-4124, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Tiwari S, Paul BN, Kumar S, Chandra A, Dhananjai S, Negi MP. Over expression of resistin in adipose tissue of the obese induces insulin resistance. World J Diabetes 2012; 3:135-41. [PMID: 22816026 PMCID: PMC3399912 DOI: 10.4239/wjd.v3.i7.135] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 05/28/2012] [Accepted: 06/10/2012] [Indexed: 02/05/2023] Open
Abstract
AIM: To compare resistin mRNA expression in subcutaneous adipose tissue (SAT) and its correlation with insulin resistance (IR) in postmenopausal obese women.
METHODS: A total of 68 postmenopausal women (non obese = 34 and obese = 34) were enrolled for the study. The women of the two groups were age matched (49-70 years). Fasting blood samples were collected at admission and abdominal SAT was obtained during surgery for gall bladder stones or hysterectomy. Physical parameters [age, height, weight, body mass index (BMI)] were measured. Biochemical (plasma insulin and plasma glucose) parameters were estimated by enzymatic methods. RNA was isolated by the Trizol method. SAT resistin mRNA expression was done by real time- reverse transcription polymerase chain reaction (RT-PCR) by using Quanti Tect SYBR Green RT-PCR master mix. Data was analyzed using independent Student’s t test, correlation and simple linear regression analysis.
RESULTS: The mean weight (52.81 ± 8.04 kg vs 79.56 ± 9.91 kg; P < 0.001), BMI (20.23 ± 3.05 kg/m2vs 32.19 ± 4.86 kg/m2; P < 0.001), insulin (8.47 ± 3.24 μU/mL vs 14.67 ± 2.18 μU/mL; P < 0.001), glucose (97.44 ± 11.31 mg/dL vs 109.67 ± 8.02 mg/dL; P < 0.001) and homeostasis model assessment index (2.01 ± 0.73 vs 3.96 ± 0.61; P < 0.001) were significantly higher in postmenopausal obese women compared to postmenopausal non obese women. The mean serum resistin level was also significantly higher in postmenopausal obese women compared to postmenopausal non obese women (9.05 ± 5.15 vs 13.92 ± 6.32, P < 0.001). Furthermore, the mean SAT resistin mRNA expression was also significantly (0.023 ± 0.008 vs 0.036 ± 0.009; P < 0.001) higher and over expressed 1.62 fold (up-regulated) in postmenopausal obese women compared to postmenopausal non obese women. In postmenopausal obese women, the relative SAT resistin mRNA expression showed positive (direct) and significant correlation with BMI (r = 0.78, P < 0.001) and serum resistin (r = 0.76, P < 0.001). Furthermore, the SAT resistin mRNA expression in postmenopausal obese women also showed significant and direct association (r = 0.45, P < 0.01) with IR, while in postmenopausal non obese women it did not show any association (r = -0.04, P > 0.05).
CONCLUSION: Increased SAT resistin mRNA expression probably leads to inducing insulin resistance and thus may be associated with obesity-related disorders in postmenopausal obese women.
Collapse
|
31
|
Nanda S, Poon LCY, Muhaisen M, Acosta IC, Nicolaides KH. Maternal serum resistin at 11 to 13 weeks' gestation in normal and pathological pregnancies. Metabolism 2012; 61:699-705. [PMID: 22146093 DOI: 10.1016/j.metabol.2011.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/09/2011] [Accepted: 10/10/2011] [Indexed: 11/24/2022]
Abstract
The objective was to examine maternal serum levels of resistin at 11 to 13 weeks' gestation in normal and pathological pregnancies. Serum resistin, pregnancy-associated plasma protein A (PAPP-A), and uterine artery pulsatility index (PI) at 11 to 13 weeks were measured in 480 singleton pregnancies, including 240 with normal outcome, 60 that subsequently developed preeclampsia (PE), 60 that developed gestational diabetes mellitus (GDM), 60 that delivered large for gestational age (LGA) neonates, and 60 that delivered small for gestational age (SGA) neonates. Each value in both the normal and pathological outcome groups was expressed as a multiple of the expected normal median (MoM), and the median MoM values in the outcome groups were compared. In the PE group, compared with the controls, there were an increase in median resistin (1.22 MoM, P = .003) and uterine artery PI (1.25 MoM, P < .0001) and a decrease in serum PAPP-A (0.72, P < .0001). There was no significant association between serum resistin with either uterine artery PI (P = .415) or serum PAPP-A (P = .290). In the SGA, LGA, and GDM groups, serum resistin MoM was not significantly different from that of the controls (P = .415, P = .702, and P = .549, respectively). In pregnancies that develop PE, maternal serum resistin concentration at 11 to 13 weeks is increased in a manner not related to altered placental perfusion or function. In pregnancies complicated by the development of GDM or delivery of SGA or LGA neonates, serum resistin is not significantly altered.
Collapse
Affiliation(s)
- Surabhi Nanda
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, UK
| | | | | | | | | |
Collapse
|
32
|
Iron in fatty liver and in the metabolic syndrome: a promising therapeutic target. J Hepatol 2011; 55:920-32. [PMID: 21718726 DOI: 10.1016/j.jhep.2011.05.008] [Citation(s) in RCA: 265] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 05/29/2011] [Accepted: 05/31/2011] [Indexed: 12/17/2022]
Abstract
The dysmetabolic iron overload syndrome (DIOS) is now a frequent finding in the general population, as is detected in about one third of patients with nonalcoholic fatty liver disease (NAFLD) and the metabolic syndrome. The pathogenesis is related to altered regulation of iron transport associated with steatosis, insulin resistance, and subclinical inflammation, often in the presence of predisposing genetic factors. Evidence is accumulating that excessive body iron plays a causal role in insulin resistance through still undefined mechanisms that probably involve a reduced ability to burn carbohydrates and altered function of adipose tissue. Furthermore, DIOS may facilitate the evolution to type 2 diabetes by altering beta-cell function, the progression of cardiovascular disease by contributing to the recruitment and activation of macrophages within arterial lesions, and the natural history of liver disease by inducing oxidative stress in hepatocytes, activation of hepatic stellate cells, and malignant transformation by promotion of cell growth and DNA damage. Based on these premises, the association among DIOS, metabolic syndrome, and NAFLD is being investigated as a new risk factor to predict the development of overt cardiovascular and hepatic diseases, and possibly hepatocellular carcinoma, but most importantly, represents also a treatable condition. Indeed, iron depletion, most frequently achieved by phlebotomy, has been shown to decrease metabolic alterations and liver enzymes in controlled studies in NAFLD. Additional studies are warranted to evaluate the potential of iron reductive therapy on hard clinical outcomes in patients with DIOS.
Collapse
|
33
|
Mazaki-Tovi S, Kanety H, Pariente C, Hemi R, Yissachar E, Schiff E, Cohen O, Sivan E. Insulin sensitivity in late gestation and early postpartum period: the role of circulating maternal adipokines. Gynecol Endocrinol 2011; 27:725-31. [PMID: 21714696 DOI: 10.3109/09513590.2010.500426] [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: 12/28/2022] Open
Abstract
OBJECTIVE Early postpartum period is characterised by a dramatic decrease in insulin resistance and significant metabolic alterations. The aims of this study were to determine the changes in circulating maternal concentrations of total adiponectin, adiponectin multimers, leptin and resistin before and after the delivery and to explore their relationship with insulin sensitivity. METHODS Twenty-seven normal pregnant women at term were included in this longitudinal study. Blood samples were taken before and 4 days after elective caesarean section. Total adiponectin, adiponectin multimers, leptin, resistin, glucose, insulin and prolactin were measured in maternal serum. Adiponectin multimers were measured before and after the delivery in eight women. RESULTS (1) The mean maternal serum total adiponectin concentration was significantly higher before than after delivery while the relative distribution of circulating maternal adiponectin multimers did not change after delivery; (2) the median maternal serum concentration of leptin was significantly higher in the antepartum than in the postpartum period; (3) the median maternal serum resistin concentration was comparable before and after delivery; (4) multiple linear regression analysis revealed that antepartum insulin sensitivity was associated with maternal low body mass index, and low glucose concentrations in glucose challenge test, as well as with maternal age and increased leptin concentrations. Postpartum insulin sensitivity was associated with decreased circulating resistin concentrations. CONCLUSIONS Despite increase in insulin sensitivity, early postpartum period is characterised by a decrease in maternal circulating total adiponectin and by steady concentrations of resistin and adiponectin multimers compared to the late third trimester.
Collapse
Affiliation(s)
- Shali Mazaki-Tovi
- Department of Obstetrics and Gynecology, Sheba Medical Center, Tel-Hashomer, Israel
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Adipokines (adipose tissue cytokines) are polypeptide factors secreted by adipose tissue in a highly regulated manner. The 'classical' adipokines (leptin, adiponectin, and resistin) are expressed only by adipocytes, but other adipokines have been shown to be released by resident and infiltrating macrophages, as well as by components of the vascular stroma. Indeed, adipose tissue inflammation is known to be associated with a modification in the pattern of adipokine secretion. Several studies indicate that adipokines can interfere with hepatic injury associated with fatty infiltration, differentially modulating steatosis, inflammation, and fibrosis. Moreover, plasma levels of adipokines have been investigated in patients with nonalcoholic fatty liver disease in order to establish correlations with the underlying state of insulin resistance and with the type and severity of hepatic damage. In this Forum article, we provide a review of recent data that suggest a significant role for oxidative stress, reactive oxygen species, and redox signaling in mediating actions of adipokines that are relevant in the pathogenesis of nonalcoholic fatty liver disease, including hepatic insulin resistance, inflammation, and fibrosis.
Collapse
Affiliation(s)
- Maurizio Parola
- Dipartimento di Medicina e Oncologia Sperimentale and Centro Interuniversitario di Fisiopatologia Epatica Università degli Studi di Torino, Turin, Italy
| | | |
Collapse
|
35
|
Heritability of genetic variants of resistin gene in patients with coronary artery disease: A family-based study. Clin Biochem 2011; 44:618-22. [DOI: 10.1016/j.clinbiochem.2011.02.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 01/29/2011] [Accepted: 02/23/2011] [Indexed: 12/20/2022]
|
36
|
Mostafaie N, Sebesta C, Zehetmayer S, Jungwirth S, Huber KR, Hinterberger M, Leitha T, Hofman J, Hejtman M, Schrattbauer K, Krugluger W, Tragl KH, Fischer P. Circulating retinol-binding protein 4 and metabolic syndrome in the elderly. Wien Med Wochenschr 2011; 161:505-10. [PMID: 21442217 DOI: 10.1007/s10354-011-0885-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Accepted: 01/12/2011] [Indexed: 11/30/2022]
Abstract
BACKGROUND Retinol-binding protein (RBP) 4, a human adipokine that specifically binds to retinol, has been reported to provide a link between obesity and insulin resistance. Plasma RBP4 concentration may be under the influence of age and obesity, but only a few studies has investigated this link in elderly individuals. Consequently, we tested the correlation between RBP4 concentrations and type 2 diabetes/metabolic syndrome (MetS) components in a large population based cohort study (VITA) of elderly [1, 2]. Using a single birth cohort, this investigation could exclude the influence of age. METHODS We evaluated the correlation of RBP4 with type 2 diabetes and MetS components including Body Mass Index (BMI), blood pressure, lipid parameters, fasting glucose insulin, homeostasis model assessment insulin resistance (HOMA-IR), and smoking in exclusively 75-76 year old participants (N = 232). RESULTS In the present study, RBP4 concentrations were associated with type 2 diabetes and metabolic syndrome (MetS) components. Of all the individual components of metabolic syndrome that were associated with RBP4 concentrations, the correlations of RBP4 with serum triglycerides and a negative correlation with HDL were the strongest ones observed in our study cohort (p<0.0001). CONCLUSIONS RBP4 plays a role in biological mechanisms that are responsible for insulin resistance and development of type 2 diabetes.
Collapse
Affiliation(s)
- Nazanin Mostafaie
- Department of Clinical Chemistry and Laboratory Medicine, Sozialmedizinisches Zentrum Ost, Danube Hospital, Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Baker R, Dauner JG, Rodriguez AC, Williams MC, Kemp TJ, Hildesheim A, Pinto LA. Increased plasma levels of adipokines and inflammatory markers in older women with persistent HPV infection. Cytokine 2011; 53:282-5. [PMID: 21167737 PMCID: PMC3033991 DOI: 10.1016/j.cyto.2010.11.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 10/20/2010] [Accepted: 11/19/2010] [Indexed: 11/23/2022]
Abstract
We observed diminished lymphoproliferation to multiple stimuli in older women with persistent cervical human papillomavirus (HPV) infection. Adipokines are a class of inflammatory cytokines that are altered in some persistent infections. The objective was to compare the level of adipokines and inflammatory cytokines in heparinized plasma from women with persistent HPV cervical infection (Cases, N=50, oversampled for their weak lymphoproliferation responses) with women with no evidence of persistent HPV cervical infection (Controls, N=50, oversampled for their strong lymphoproliferation responses). Plasma samples were analyzed with multiplex assays for adipokines and inflammatory cytokines. Cases had significantly elevated plasma levels of resistin (p<0.0001) and sFas (p=0.0038) as compared to controls. Risk of persistent HPV infection increased significantly with increasing levels of resistin and 8Fas. This is the first study to demonstrate elevated levels of resistin and sFas in HPV persistently infected, older women with decreased immune function expanding the understanding of the systemic inflammation and immune alterations in individuals persistently infected with HPV. Further studies within a larger cohort are needed to define the generalities of these findings and any role adipokines have in persistent HPV infection.
Collapse
Affiliation(s)
- Rosalyn Baker
- HPV Immunology Laboratory, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 21702, USA
- Division of Clinical Research/ICMOB, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joseph G. Dauner
- HPV Immunology Laboratory, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 21702, USA
| | | | - Marcus C. Williams
- HPV Immunology Laboratory, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 21702, USA
| | - Troy J. Kemp
- HPV Immunology Laboratory, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 21702, USA
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ligia A. Pinto
- HPV Immunology Laboratory, SAIC-Frederick, Inc., NCI-Frederick, Frederick, MD, 21702, USA
| |
Collapse
|
38
|
Correlation of plasma resistin with obesity and insulin resistance in type 2 diabetic patients. DIABETES & METABOLISM 2010; 36:443-9. [PMID: 20739208 DOI: 10.1016/j.diabet.2010.05.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 05/20/2010] [Accepted: 05/26/2010] [Indexed: 12/17/2022]
|
39
|
Gupta V, Singh A, Pant A. Could resistin be a noble marker for metabolic syndrome? Diabetes & Metabolic Syndrome: Clinical Research & Reviews 2010. [DOI: 10.1016/j.dsx.2010.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
40
|
Osawa H, Tabara Y, Ohashi J, Kawamura R, Onuma H, Makino H. Is rs34861192 or rs1862513 a more promising variant for determining plasma resistin in an aged Japanese population? Diabetologia 2010; 53:795-7. [PMID: 20131043 PMCID: PMC2830588 DOI: 10.1007/s00125-010-1665-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 12/07/2009] [Indexed: 11/30/2022]
Affiliation(s)
- H. Osawa
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295 Japan
- Ehime Proteo-Medicine Research Center, Ehime University, Toon, Ehime Japan
| | - Y. Tabara
- Department of Basic Medical Research and Education, Ehime University Graduate School of Medicine, Toon, Ehime Japan
- Ehime Proteo-Medicine Research Center, Ehime University, Toon, Ehime Japan
| | - J. Ohashi
- Doctoral Program in Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
| | - R. Kawamura
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295 Japan
| | - H. Onuma
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295 Japan
- Ehime Proteo-Medicine Research Center, Ehime University, Toon, Ehime Japan
| | - H. Makino
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295 Japan
- Institute of Diabetes Research Center, Takanoko Hospital, Matsuyama, Ehime Japan
| |
Collapse
|
41
|
Onuma H, Tabara Y, Kawamura R, Tanaka T, Ohashi J, Nishida W, Takata Y, Ochi M, Yamada K, Kawamoto R, Kohara K, Miki T, Makino H, Osawa H. A at single nucleotide polymorphism-358 is required for G at -420 to confer the highest plasma resistin in the general Japanese population. PLoS One 2010; 5:e9718. [PMID: 20300528 PMCID: PMC2838794 DOI: 10.1371/journal.pone.0009718] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 02/14/2010] [Indexed: 12/20/2022] Open
Abstract
Insulin resistance is a feature of type 2 diabetes. Resistin, secreted from adipocytes, causes insulin resistance in mice. We previously reported that the G/G genotype of single nucleotide polymorphism (SNP) at −420 (rs1862513) in the human resistin gene (RETN) increased susceptibility to type 2 diabetes by enhancing its promoter activity. Plasma resistin was highest in Japanese subjects with G/G genotype, followed by C/G, and C/C. In this study, we cross-sectionally analyzed plasma resistin and SNPs in the RETN region in 2,019 community-dwelling Japanese subjects. Plasma resistin was associated with SNP-638 (rs34861192), SNP-537 (rs34124816), SNP-420, SNP-358 (rs3219175), SNP+299 (rs3745367), and SNP+1263 (rs3745369) (P<10−13 in all cases). SNP-638, SNP -420, SNP-358, and SNP+157 were in the same linkage disequilibrium (LD) block. SNP-358 and SNP-638 were nearly in complete LD (r2 = 0.98), and were tightly correlated with SNP-420 (r2 = 0.50, and 0.51, respectively). The correlation between either SNP-358 (or SNP-638) or SNP-420 and plasma resistin appeared to be strong (risk alleles for high plasma resistin; A at SNP-358, r2 = 0.5224, P = 4.94×10−324; G at SNP-420, r2 = 0.2616, P = 1.71×10−133). In haplotypes determined by SNP-420 and SNP-358, the estimated frequencies for C-G, G-A, and G-G were 0.6700, 0.2005, and 0.1284, respectively, and C-A was rare (0.0011), suggesting that subjects with A at −358, generally had G at −420. This G-A haplotype conferred the highest plasma resistin (8.24 ng/ml difference/allele compared to C-G, P<0.0001). In THP-1 cells, the RETN promoter with the G-A haplotype showed the highest activity. Nuclear proteins specifically recognized one base difference at SNP-358, but not at SNP-638. Therefore, A at -358 is required for G at −420 to confer the highest plasma resistin in the general Japanese population. In Caucasians, the association between SNP-420 and plasma resistin is not strong, and A at −358 may not exist, suggesting that SNP-358 could explain this ethnic difference.
Collapse
Affiliation(s)
- Hiroshi Onuma
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
- Ehime Proteo-Medicine Research Center, Ehime University, Ehime, Japan
| | - Yasuharu Tabara
- Ehime Proteo-Medicine Research Center, Ehime University, Ehime, Japan
- Department of Basic Medical Research and Education, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryoichi Kawamura
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Takashi Tanaka
- Laboratory of Molecular Biology, Faculty of Pharmacy, Osaka Ohtani University, Osaka, Japan
| | - Jun Ohashi
- Doctoral Program in Life System Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Wataru Nishida
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Yasunori Takata
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masaaki Ochi
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kazuya Yamada
- Department of Health and Nutritional Science, Faculty of Human Health Science, Matsumoto University, Nagano, Japan
| | - Ryuichi Kawamoto
- Department of Community Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Katsuhiko Kohara
- Ehime Proteo-Medicine Research Center, Ehime University, Ehime, Japan
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Tetsuro Miki
- Ehime Proteo-Medicine Research Center, Ehime University, Ehime, Japan
- Department of Geriatric Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hideichi Makino
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Haruhiko Osawa
- Department of Molecular and Genetic Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
- Ehime Proteo-Medicine Research Center, Ehime University, Ehime, Japan
- * E-mail:
| |
Collapse
|
42
|
Menzaghi C, Trischitta V. Genetics of serum resistin: a paradigm of population-specific regulation? Diabetologia 2010; 53:226-8. [PMID: 19882136 DOI: 10.1007/s00125-009-1589-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 10/05/2009] [Indexed: 10/20/2022]
Affiliation(s)
- C Menzaghi
- Research Unit of Diabetes and Endocrine Diseases, IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013 San Giovanni Rotondo, FG, Italy.
| | | |
Collapse
|
43
|
Asano H, Izawa H, Nagata K, Nakatochi M, Kobayashi M, Hirashiki A, Shintani S, Nishizawa T, Tanimura D, Naruse K, Matsubara T, Murohara T, Yokota M. Plasma resistin concentration determined by common variants in the resistin gene and associated with metabolic traits in an aged Japanese population. Diabetologia 2010; 53:234-46. [PMID: 19727657 DOI: 10.1007/s00125-009-1517-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2009] [Accepted: 08/05/2009] [Indexed: 01/22/2023]
Abstract
AIMS/HYPOTHESIS Resistin is a cytokine derived from adipose tissue and is implicated in obesity-related insulin resistance and type 2 diabetes mellitus. Polymorphisms of the resistin gene (RETN) have been shown to affect the plasma resistin concentration. The aims of this study were to identify polymorphisms of RETN that influence plasma resistin concentration and to clarify the relation between plasma resistin level and metabolic disorders in an aged Japanese cohort. METHODS The study participants comprised 3133 individuals recruited to a population-based prospective cohort study (KING study). Plasma resistin concentration, BMI, abdominal circumference, blood pressure, fasting plasma glucose and serum insulin concentrations, HbA(1c) content and serum lipid profile were measured in all participants. The HOMA index of insulin resistance (HOMA-IR) was also calculated. Eleven polymorphisms of RETN were genotyped. RESULTS A combination of ANOVA and multiple linear regression analysis in screening and large-scale subsets of the study population revealed that plasma resistin concentration was significantly associated with rs34861192 and rs3745368 polymorphisms of RETN. Multiple linear regression analysis with adjustment for age and sex also showed that the plasma resistin level was significantly associated with serum concentrations of HDL-cholesterol, triacylglycerol and insulin, as well as with BMI. CONCLUSIONS/INTERPRETATION Our results implicate the rs34861192 and rs3745368 polymorphisms of RETN as robust and independent determinants of plasma resistin concentration in the study population. In addition, plasma resistin level was associated with dyslipidaemia, serum insulin concentration and obesity. TRIAL REGISTRATION ClinicalTrials.gov NCT00262691.
Collapse
Affiliation(s)
- H Asano
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pasarica M, Gowronska-Kozak B, Burk D, Remedios I, Hymel D, Gimble J, Ravussin E, Bray GA, Smith SR. Adipose tissue collagen VI in obesity. J Clin Endocrinol Metab 2009; 94:5155-62. [PMID: 19837927 PMCID: PMC2819828 DOI: 10.1210/jc.2009-0947] [Citation(s) in RCA: 232] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Basic science studies show that the extracellular matrix of adipose tissue, mainly represented by collagen VI, is dysfunctional in obesity and contributes to the development of the metabolic syndrome. We hypothesized in humans that increased collagen VI alpha3-subunit (COL6A3) mRNA is associated with adipose tissue macrophage chemotaxis and inflammation and that weight gain is accompanied by changes in the expression of COL6A3. RESEARCH DESIGN AND METHODS Adipose tissue biopsies were obtained from a cross-sectional study (n = 109), an overfeeding study (n = 9), and a pioglitazone treatment study (n = 14). Adipose tissue gene expression was measured by quantitative RT-PCR, immunohistochemistry, and adipocyte sizing by fixation with osmium and Coulter counting. Body composition was measured by dual-energy x-ray absorptiometry and visceral adipose tissue by computed tomography. Patients with high or low COL6A3 mRNA were compared by one-way ANOVA. RESULTS In humans, immunohistochemistry revealed that COL6 is present in adipose tissue extracellular matrix. COL6A3 mRNA is correlated with body mass index (r = 0.60, P < 0.0001) and fat mass (r = 0.41, P < 0.0001). COL6A3 expression was similar in obese vs. type 2 diabetes patients. Obese subjects with high COL6A3 mRNA had greater visceral adipose tissue mass (P < 0.05), lower size of small and medium adipocytes (P < 0.05), more CD68+ and CD163/MAC2+ macrophages, and increased macrophage inflammatory protein-1alpha and macrophage chemoattractant protein-1alpha mRNA (P < 0.05). Eight weeks of overfeeding increased body weight and COL6A3 mRNA (P < 0.05). Pioglitazone decreased COL6A3 mRNA, and the change was inversely proportional to baseline COL6A3 mRNA (r = -0.95, P < 0.0001). CONCLUSION These results are consistent with basic science data, suggesting that COL6A3 might contribute to adipose tissue inflammation.
Collapse
Affiliation(s)
- Magdalena Pasarica
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, Louisiana 70808, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Singh PP, Mahadi F, Roy A, Sharma P. Reactive oxygen species, reactive nitrogen species and antioxidants in etiopathogenesis of diabetes mellitus type-2. Indian J Clin Biochem 2009; 24:324-42. [PMID: 23105858 PMCID: PMC3453064 DOI: 10.1007/s12291-009-0062-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus type-2 (DMT-2) is a hyperglycemic syndrome with several characteristic features. It continues to rise unabatedly in all pockets of the world, parallels with affluence and can be controlled but not cured. It has a definite involvement of genetic component but environmental factors play overwhelmingly dominant role in etiopathogenesis. Insulin resistance (IR) and obesity are singular instigators of DMT-2. The various events cause critical defects in insulin signaling cascade followed by beta-cell dysfunction. Over a period of time, numerous other metabolic aberrations develop, resulting in diabetic complications which could be both vascular (cardiovascular complications, nephropathy, neuropathy, retinopathy and embryopathy) or a-vascular (cataract and glaucoma etc). It has been proposed that all these abnormal events are initiated or activated by a common mechanism of superoxide anion, which is accompanied with generation of a variety of reactive oxygen species (ROS), reactive nitrogen specie (RNS) and resultant heightened oxidative stress (OS). Provoked OS causes IR and altered gene expressions. Hyperglycemia induces OS through multiple routes: a)stimulated polyol pathway where in ≤ 30% glucose can be diverted to sorbitol and fructose, b)increased transcription of genes for proinflammatory cytokines and plasminogen activator inhibitor-1 (PAI-1) c) activation of protein kinase-C (PKC) leading to several molecular changes d)increased synthesis of Advanced Glycation End Products (AGEs) e)changes in a receptor far AGEs and f) autooxidation of glucose with formation of ketoimines and AGEs. All these processes are accompanied with alteration in redox status, ROS, RNS and OS which trigger DMT-2 and its complications. Initial hurriedly planned and executed experimental and clinical studies showed promising results of antioxidant therapies, but recent studies indicate that excess intake/supplement may have adverse outcomes including increased mortality. It is advocated that antioxidants should be given only if preexisting deficiency is present. Selection of antioxidant is another important aspect. Lastly but most importantly the impact of OS is not obligatory but facultative. As such only those diabetic patients will be benefited by antioxidant therapies that have impelling punch of prooxidants.
Collapse
Affiliation(s)
- P. P. Singh
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, Uttar Pradesh India
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, 226003 India
| | - Farzana Mahadi
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, Uttar Pradesh India
| | - Ajanta Roy
- Department of Biochemistry, Era’s Lucknow Medical College, Sarfarazganj, Lucknow, Uttar Pradesh India
| | - Praveen Sharma
- Department of Biochemistry, SMS Medical College Jaipur, Rajasthan, India
| |
Collapse
|
46
|
Luo Z, Zhang Y, Li F, He J, Ding H, Yan L, Cheng H. Resistin induces insulin resistance by both AMPK-dependent and AMPK-independent mechanisms in HepG2 cells. Endocrine 2009; 36:60-9. [PMID: 19440859 DOI: 10.1007/s12020-009-9198-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 03/30/2009] [Accepted: 04/08/2009] [Indexed: 12/19/2022]
Abstract
Resistin is a 12.5-KDa cysteine-rich peptide that has been implicated in the impairment of glucose homeostasis via the AMP-activated protein kinase (AMPK) pathway in a rodent model. However, the role resistin plays in humans is controversial. This study investigated the effect of resistin on glucose metabolism and insulin signaling using human recombinant resistin and small interfering RNA (siRNA) against AMPKalpha2 to treat the human liver HepG2 cells. The mRNA of key genes involved in glucose metabolism and the insulin-signaling pathway were detected by real-time RT-PCR. Phosphorylation levels of Akt and AMPK were measured by western blot. The incorporation of D-[U-(14)C] glucose into glycogen was quantitated by liquid scintillation counting. The results demonstrate that resistin stimulated expressions of glucose-6-phosphatase (G6Pase), phosphoenolypyruvate carboxykinase (PEPCK), and suppressor of cytokine signaling 3 (SOCS-3), repressed the expressions of insulin receptor substrate 2(IRS-2) and glucose transporter 2(GLUT2). In addition, resistin inhibited the insulin-induced phosphorylation of Akt independent of AMPK. In conclusion, our findings suggest that resistin induces insulin resistance in HepG2 cells at least partly via induction of SOCS-3 expression and reduction of Akt phosphorylation through an AMPK-independent mechanism. Resistin also increases glucose production via AMPK-mediated upregulated expression of the genes encoding hepatic gluconeogenic enzymes, G6Pase, and PEPCK.
Collapse
Affiliation(s)
- Zhaofan Luo
- Department of Endocrinology, The Second Affiliated Hospital, Sun Yat-Sen University, 510120, Guangzhou, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
47
|
Won JC, Park CY, Lee WY, Lee ES, Oh SW, Park SW. Association of plasma levels of resistin with subcutaneous fat mass and markers of inflammation but not with metabolic determinants or insulin resistance. J Korean Med Sci 2009; 24:695-700. [PMID: 19654955 PMCID: PMC2719220 DOI: 10.3346/jkms.2009.24.4.695] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Accepted: 08/22/2008] [Indexed: 01/28/2023] Open
Abstract
The aim of the present study was to investigate the relationship of plasma resistin levels with determinants of the metabolic syndrome (MetS) and anthropometric parameters in healthy Korean subjects. Plasma resistin levels were determined in 276 subjects. In subjects with MetS, the plasma resistin levels were not significantly increased compared to those without MetS (8.3+/-4.3 ng/mL vs. 8.5+/-3.6 ng/mL, respectively, P=0.84). In addition, the plasma resistin levels were not correlated with the body mass index, the waist circumference, homeostasis model assessment-insulin resistance (HOMA-IR), fasting plasma glucose or insulin levels. However, the plasma resistin levels were positively correlated with the abdominal subcutaneous fat (r=0.18, P<0.01) in all subjects and correlated with TNF alpha(r=-0.16, P<0.05) and hsCRP (r=0.15, P<0.05) in subjects without MetS but not with MetS. With multiple linear regression analysis, these linear associations remained to be significant. The results of this study show that plasma resistin levels in humans were not associated with markers of insulin resistance, obesity or other determinants of the MetS.
Collapse
Affiliation(s)
- Jong Chul Won
- Department of Internal Medicine, Sungkyunkwan University School of Medicine, Kangbuk Samsung Hospital, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
48
|
Hagiwara S, Gohda T, Tanimoto M, Ito T, Murakoshi M, Ohara I, Yamazaki T, Matsumoto M, Horikoshi S, Funabiki K, Tomino Y. Effects of pyridoxamine (K-163) on glucose intolerance and obesity in high-fat diet C57BL/6J mice. Metabolism 2009; 58:934-45. [PMID: 19427656 DOI: 10.1016/j.metabol.2009.02.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Accepted: 02/19/2009] [Indexed: 01/11/2023]
Abstract
Advanced glycation end products (AGEs) contribute to the pathogenesis of diabetes-associated complications. Previously, we reported the possible effect of pyridoxamine (K-163), an AGE inhibitor, on improvement of glucose intolerance in type 2 diabetes mellitus KK-A(y)/Ta mice. Recently, AGEs and oxidative stress have been shown to induce insulin resistance. The objective of the present study is to examine the effect of pyridoxamine on glucose intolerance and oxidative stress. C57BL/6J mice were divided into 3 groups as follows: low-fat diet, high-fat diet, and high-fat diet with pyridoxamine treatment. Body and adipose tissue weight, serum insulin, hydrogen peroxide, malondialdehyde and AGE, and urinary 8-hydroxy-2'-deoxyguanosine levels were measured. Nicotinamide adenine dinucleotide phosphate subunits, antioxidant enzymes, and adipocytokine messenger RNA expressions in the adipose tissues were evaluated. Akt/protein kinase B activity and glucose transporter 4 translocation in skeletal muscle were also evaluated. Body and adipose tissue weights of the pyridoxamine treatment group were significantly decreased compared with those of the high-fat diet group. Pyridoxamine attenuated serum hydrogen peroxide, malondialdehyde and AGE, and urinary 8-hydroxy-2'-deoxyguanosine and nicotinamide adenine dinucleotide phosphate oxidase expression; increased antioxidant enzyme expression; and improved dysregulation of adipocytokines in adipose tissues. Pyridoxamine improved blood glucose levels after glucose injection and fasting hyperinsulinemia. Suppressed Akt/protein kinase B activity and glucose transporter 4 translocation in skeletal muscle in high-fat diet mice were improved by pyridoxamine treatment. It appears that the antioxidative effect of pyridoxamine is associated with improvement of glucose intolerance and obesity in C57BL/6J mice fed a high-fat diet. We assume that pyridoxamine may be useful in the treatment of the obesity-associated metabolic syndrome.
Collapse
Affiliation(s)
- Shinji Hagiwara
- Department of Internal Medicine, Division of Nephrology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Wågsäter D, Mumtaz M, Löfgren S, Hugander A, Dimberg J. Resistin in Human Colorectal Cancer: Increased Expression Independently of Resistin Promoter −420C > G genotype. Cancer Invest 2009; 26:1008-14. [DOI: 10.1080/07357900802087267] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
50
|
Pechlivanis S, Bermejo JL, Pardini B, Naccarati A, Vodickova L, Novotny J, Hemminki K, Vodicka P, Försti A. Genetic variation in adipokine genes and risk of colorectal cancer. Eur J Endocrinol 2009; 160:933-40. [PMID: 19273568 DOI: 10.1530/eje-09-0039] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Obesity has been related to an increased risk of colorectal cancer (CRC). Adipokines produced by the adipose tissue are directly linked to obesity and may thus contribute to the pathogenesis of CRC. We hypothesized that potentially functional polymorphisms in the adipokine genes leptin (LEP), leptin receptor (LEPR), resistin (RETN), and adiponectin (ADIPOQ) may be associated with CRC. DESIGN AND METHODS We studied the association of four putatively functional single nucleotide polymorphisms (SNPs) with CRC risk using a hospital-based study design with 702 cases and 752 controls from the Czech Republic. We used likelihood ratio tests to select the best model to represent the relationship between genotypes and risk of CRC. Age-adjusted odds ratios (ORs) under the best model were calculated for each SNP. Previous genotyping data on insulin (INS)-related genes were used to explore interactions between genes in obesity- and diabetes-related pathways by using two independent methods, logistic regression, and multifactor-dimensionality reduction. RESULTS A trend to associate between the RETN SNP rs1862513 (C-420G) and CRC risk was observed (per allele OR 1.18, 95% confidence interval (0.99-1.40). Statistically, significant interactions were observed between the INS SNP rs3842754 (+1127INSPstI) genotypes and both the LEPR SNP rs1137101 (Q223R) and the ADIPOQ SNP rs266729 (C-11374G) genotypes. CONCLUSIONS Our results suggest that variants in the adipokine genes may affect CRC risk in combination with variants in diabetes-related genes.
Collapse
Affiliation(s)
- Sonali Pechlivanis
- Division of Molecular Genetic Epidemiology C050, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|