1
|
Ivovic A, Yung JHM, Oprescu AI, Vlavcheski F, Mori Y, Rahman SMN, Ye W, Eversley JA, Wheeler MB, Woo M, Tsiani E, Giacca A. β-Cell Insulin Resistance Plays a Causal Role in Fat-Induced β-Cell Dysfunction In Vitro and In Vivo. Endocrinology 2024; 165:bqae044. [PMID: 38578954 PMCID: PMC11033845 DOI: 10.1210/endocr/bqae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 04/07/2024]
Abstract
In the classical insulin target tissues of liver, muscle, and adipose tissue, chronically elevated levels of free fatty acids (FFA) impair insulin signaling. Insulin signaling molecules are also present in β-cells where they play a role in β-cell function. Therefore, inhibition of the insulin/insulin-like growth factor 1 pathway may be involved in fat-induced β-cell dysfunction. To address the role of β-cell insulin resistance in FFA-induced β-cell dysfunction we co-infused bisperoxovanadate (BPV) with oleate or olive oil for 48 hours in rats. BPV, a tyrosine phosphatase inhibitor, acts as an insulin mimetic and is devoid of any antioxidant effect that could prevent β-cell dysfunction, unlike most insulin sensitizers. Following fat infusion, rats either underwent hyperglycemic clamps for assessment of β-cell function in vivo or islets were isolated for ex vivo assessment of glucose-stimulated insulin secretion (GSIS). We also incubated islets with oleate or palmitate and BPV for in vitro assessment of GSIS and Akt (protein kinase B) phosphorylation. Next, mice with β-cell specific deletion of PTEN (phosphatase and tensin homolog; negative regulator of insulin signaling) and littermate controls were infused with oleate for 48 hours, followed by hyperglycemic clamps or ex vivo evaluation of GSIS. In rat experiments, BPV protected against fat-induced impairment of β-cell function in vivo, ex vivo, and in vitro. In mice, β-cell specific deletion of PTEN protected against oleate-induced β-cell dysfunction in vivo and ex vivo. These data support the hypothesis that β-cell insulin resistance plays a causal role in FFA-induced β-cell dysfunction.
Collapse
Affiliation(s)
- Aleksandar Ivovic
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Justin Hou Ming Yung
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Andrei I Oprescu
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Filip Vlavcheski
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Yusaku Mori
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Diabetes, Metabolism, and Endocrinology, Anti-Glycation Research Section, Department of Medicine, Showa University School of Medicine, Shinagawa, Tokyo 142-8555, Japan
| | - S M Niazur Rahman
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Wenyue Ye
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Judith A Eversley
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Minna Woo
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Toronto General Hospital Research Institute, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
- Division of Endocrinology, Department of Medicine, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
2
|
Bouchard-Mercier A, de Toro-Martín J, Nadeau M, Lescelleur O, Lebel S, Richard D, Biertho L, Tchernof A, Vohl MC. Molecular remodeling of adipose tissue is associated with metabolic recovery after weight loss surgery. J Transl Med 2022; 20:283. [PMID: 35739539 PMCID: PMC9219157 DOI: 10.1186/s12967-022-03485-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/13/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Bariatric surgery is an effective therapy for individuals with severe obesity to achieve sustainable weight loss and to reduce comorbidities. Examining the molecular signature of subcutaneous adipose tissue (SAT) following different types of bariatric surgery may help in gaining further insight into their distinct metabolic impact. RESULTS Subjects undergoing biliopancreatic diversion with duodenal switch (BPD-DS) showed a significantly higher percentage of total weight loss than those undergoing gastric bypass or sleeve gastrectomy (RYGB + SG) (41.7 ± 4.6 vs 28.2 ± 6.8%; p = 0.00005). Individuals losing more weight were also significantly more prone to achieve both type 2 diabetes and dyslipidemia remission (OR = 0.75; 95%CI = 0.51-0.91; p = 0.03). Whole transcriptome and methylome profiling showed that bariatric surgery induced a profound molecular remodeling of SAT at 12 months postoperative, mainly through gene down-regulation and hypermethylation. The extent of changes observed was greater following BPD-DS, with 61.1% and 49.8% of up- and down-regulated genes, as well as 85.7% and 70.4% of hyper- and hypomethylated genes being exclusive to this procedure, and mostly associated with a marked decrease of immune and inflammatory responses. Weight loss was strongly associated with genes being simultaneously differentially expressed and methylated in BPD-DS, with the strongest association being observed for GPD1L (r2 = 0.83; p = 1.4 × 10-6). CONCLUSIONS Present findings point to the greater SAT molecular remodeling following BPD-DS as potentially linked with higher metabolic remission rates. These results will contribute to a better understanding of the metabolic pathways involved in the response to bariatric surgery and will eventually lead to the development of gene targets for the treatment of obesity. Trial registration ClinicalTrials.gov NCT02390973.
Collapse
Affiliation(s)
- Annie Bouchard-Mercier
- School of Nutrition and Centre Nutrition, Santé et Société (NUTRISS)-Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Pavillon des Services (suite 2729K), 2440 Hochelaga Blvd, Quebec City, QC G1V 0A6 Canada
| | - Juan de Toro-Martín
- School of Nutrition and Centre Nutrition, Santé et Société (NUTRISS)-Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Pavillon des Services (suite 2729K), 2440 Hochelaga Blvd, Quebec City, QC G1V 0A6 Canada
| | - Mélanie Nadeau
- Centre de recherche de l’institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 chemin Sainte-Foy, Quebec City, QC G1V 4G5 Canada
| | - Odette Lescelleur
- Centre de recherche de l’institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 chemin Sainte-Foy, Quebec City, QC G1V 4G5 Canada
| | - Stéfane Lebel
- Centre de recherche de l’institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 chemin Sainte-Foy, Quebec City, QC G1V 4G5 Canada
| | - Denis Richard
- Centre de recherche de l’institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 chemin Sainte-Foy, Quebec City, QC G1V 4G5 Canada
| | - Laurent Biertho
- Centre de recherche de l’institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 chemin Sainte-Foy, Quebec City, QC G1V 4G5 Canada
| | - André Tchernof
- Centre de recherche de l’institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 chemin Sainte-Foy, Quebec City, QC G1V 4G5 Canada
| | - Marie-Claude Vohl
- School of Nutrition and Centre Nutrition, Santé et Société (NUTRISS)-Institut sur la nutrition et les aliments fonctionnels (INAF), Université Laval, Pavillon des Services (suite 2729K), 2440 Hochelaga Blvd, Quebec City, QC G1V 0A6 Canada
| |
Collapse
|
3
|
Timmons JA, Anighoro A, Brogan RJ, Stahl J, Wahlestedt C, Farquhar DG, Taylor-King J, Volmar CH, Kraus WE, Phillips SM. A human-based multi-gene signature enables quantitative drug repurposing for metabolic disease. eLife 2022; 11:68832. [PMID: 35037854 PMCID: PMC8763401 DOI: 10.7554/elife.68832] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 11/26/2021] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance (IR) contributes to the pathophysiology of diabetes, dementia, viral infection, and cardiovascular disease. Drug repurposing (DR) may identify treatments for IR; however, barriers include uncertainty whether in vitro transcriptomic assays yield quantitative pharmacological data, or how to optimise assay design to best reflect in vivo human disease. We developed a clinical-based human tissue IR signature by combining lifestyle-mediated treatment responses (>500 human adipose and muscle biopsies) with biomarkers of disease status (fasting IR from >1200 biopsies). The assay identified a chemically diverse set of >130 positively acting compounds, highly enriched in true positives, that targeted 73 proteins regulating IR pathways. Our multi-gene RNA assay score reflected the quantitative pharmacological properties of a set of epidermal growth factor receptor-related tyrosine kinase inhibitors, providing insight into drug target specificity; an observation supported by deep learning-based genome-wide predicted pharmacology. Several drugs identified are suitable for evaluation in patients, particularly those with either acute or severe chronic IR.
Collapse
Affiliation(s)
- James A Timmons
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Augur Precision Medicine LTD, Stirling, United Kingdom
| | | | | | - Jack Stahl
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, United States
| | - Claes Wahlestedt
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, United States
| | | | | | - Claude-Henry Volmar
- Center for Therapeutic Innovation, Miller School of Medicine, University of Miami, Miami, United States
| | | | - Stuart M Phillips
- Faculty of Science, Kinesiology, McMaster University, Hamilton, Canada
| |
Collapse
|
4
|
Ranganath Pai K, Manokaran K, Bhat P, Nayak D, Baskaran R, Paramasivam P, Ahmed S, Priya K, Balaji V. Oxidative stress and female reproductive disorder: A review. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2022. [DOI: 10.4103/2305-0500.346088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
5
|
PPARs-Orchestrated Metabolic Homeostasis in the Adipose Tissue. Int J Mol Sci 2021; 22:ijms22168974. [PMID: 34445679 PMCID: PMC8396609 DOI: 10.3390/ijms22168974] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/17/2021] [Indexed: 01/12/2023] Open
Abstract
It has been more than three decades since peroxisome proliferator-activated receptors (PPARs) were first discovered. Many investigations have revealed the central regulators of PPARs in lipid and glucose homeostasis in response to different nutrient conditions. PPARs have attracted much attention due to their ability to improve metabolic syndromes, and they have also been proposed as classical drug targets for the treatment of hyperlipidemia and type 2 diabetes (T2D) mellitus. In parallel, adipose tissue is known to play a unique role in the pathogenesis of insulin resistance and metabolic syndromes due to its ability to “safely” store lipids and secrete cytokines that regulate whole-body metabolism. Adipose tissue relies on a complex and subtle network of transcription factors to maintain its normal physiological function, by coordinating various molecular events, among which PPARs play distinctive and indispensable roles in adipocyte differentiation, lipid metabolism, adipokine secretion, and insulin sensitivity. In this review, we discuss the characteristics of PPARs with special emphasis on the roles of the different isotypes in adipocyte biology.
Collapse
|
6
|
Jiang JX, Tomilov A, Montgomery C, Hui CK, Török NJ, Cortopassi G. Shc inhibitor idebenone ameliorates liver injury and fibrosis in dietary NASH in mice. J Biochem Mol Toxicol 2021; 35:e22876. [PMID: 34369032 DOI: 10.1002/jbt.22876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 07/20/2021] [Accepted: 07/24/2021] [Indexed: 12/20/2022]
Abstract
Shc expression rises in human nonalcoholic steatohepatitis (NASH) livers, and Shc-deficient mice are protected from NASH-thus Shc inhibition could be a novel therapeutic strategy for NASH. Idebenone was recently identified as the first small-molecule Shc inhibitor drug. We tested idebenone in the fibrotic methionine-choline deficient (MCD) diet and the metabolic fast food diet (FFD) mouse models of NASH. In the fibrotic MCD NASH model, idebenone reduced Shc expression and phosphorylation in peripheral blood mononuclear cells and Shc expression in the liver; decreased serum alanine aminotransferase and aspartate aminotransferase; and attenuated liver fibrosis as observed by quantitative polymerase chain reaction (qPCR) and hydroxyproline quantification. In the metabolic FFD model, idebenone administration improved insulin resistance, and reduced inflammation and fibrosis shown with qPCR, hydroxyproline measurement, and histology. Thus, idebenone ameliorates NASH in two mouse models. As an approved drug with a benign safety profile, Idebenone could be a reasonable human NASH therapy.
Collapse
Affiliation(s)
- Joy X Jiang
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Alexey Tomilov
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Claire Montgomery
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Chun Kui Hui
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Natalie J Török
- Gastroenterology and Hepatology, Stanford University, Palo Alto, California, USA.,VA Palo Alto, Palo Alto, California, USA
| | - Gino Cortopassi
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| |
Collapse
|
7
|
Sharma M, Aggarwal S, Nayar U, Vikram NK, Misra A, Luthra K. Differential expression of insulin receptor substrate-1(IRS-1) in visceral and subcutaneous adipose depots of morbidly obese subjects undergoing bariatric surgery in a tertiary care center in north India; SNP analysis and correlation with metabolic profile. Diabetes Metab Syndr 2021; 15:981-986. [PMID: 33975152 DOI: 10.1016/j.dsx.2021.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND /aim: Abdominal obesity and associated metabolic consequences are a burgeoning problem in Asian Indians and studying their genetic predisposition is important. This study is aimed at assessing variations in Insulin receptor substrate-1 (IRS-1), its expression at regional fat-depots (visceral and subcutaneous) in morbidly obese patients, and correlation with genotype-phenotype traits. METHODS Gene expression of IRS-1 in paired adipose tissue from 35 morbidly obese subjects (BMI) > 40 kg/m2) with co-morbidities and 15 controls (BMI<25 kg/m2), undergoing bariatric/elective abdominal surgery, respectively was determined by quantitative real time PCR. Genotyping of IRS-1Gly972Arg (n = 436) (rs 1801278) was performed by PCR-RFLP. Metabolic parameters were assessed. Full length sequencing of IRS-1 was performed to identify known/novel variations. RESULTS A marked reduction in IRS-1 expression was observed in visceral as compared to subcutaneous adipose tissue of morbidly obese subjects (p = 0.02). Homozygous variant of IRS-1 Gly972Arg was absent and there was no association with obesity or insulin resistance. A salient finding of this study was identification of two new variants in IRS-1 gene, representing G > A (codon 1102) encoding Glu > Lys and a deletion of (A) at codon 658 in morbidly obese subjects with insulin resistance. CONCLUSIONS Observation of a substantially lower expression of IRS-1 for first time in visceral adipose tissue of morbidly obese subjects is suggestive of predictive role of IRS-1 expression in insulin responsiveness of visceral adipose tissue. New variants in IRS-1, a non-synonymous mutation and a deletion should be evaluated further for their role in development of obesity and/orT2DM.
Collapse
Affiliation(s)
- Mukti Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Delhi, India
| | - Sandeep Aggarwal
- Department of Surgery, All India Institute of Medical Sciences, Delhi, India
| | - Umakant Nayar
- Department of Surgery, All India Institute of Medical Sciences, Delhi, India
| | | | - Anoop Misra
- Diabetic Foundation, National Diabetes Obesity and Cholesterol Foundation (N-DOC), New Delhi, India; Fortis C-DOC Center of Excellence for Diabetes, Metabolic Diseases, and Endocrinology, B 16, Chirag Enclave, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, Delhi, India.
| |
Collapse
|
8
|
Hu J, Xu W, Yang H, Mu L. Uric acid participating in female reproductive disorders: a review. Reprod Biol Endocrinol 2021; 19:65. [PMID: 33906696 PMCID: PMC8077923 DOI: 10.1186/s12958-021-00748-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/18/2021] [Indexed: 12/21/2022] Open
Abstract
Uric acid (UA) is the end metabolic product of purine metabolism. Early on, UA was considered to be a metabolite with a certain antioxidant capacity. As research has progressed, other properties of UA have been explored, and its association with many diseases has been found. The association between UA and kidney disease and cardiovascular disease is well established; however, there is still a paucity of reviews on the association between UA and the female reproductive system. An increasing number of epidemiological studies have shown elevated serum UA levels in patients with polycystic ovary syndrome (PCOS), endometriosis, etc. Additionally, serum UA can be used as a predictor of pregnancy complications and adverse foetal outcomes. An increasing number of animal experiments and clinical studies have revealed possible mechanisms related to the involvement of UA in certain female reproductive disorders: oxidative stress, chronic inflammation, mitochondrial dysfunction, etc. This article reviews the current mainstream mechanisms regarding the pathogenesis of UA and the role of UA in certain specific female reproductive disorders (direct involvement in the development of certain diseases or enhancement of other risk factors) in the hope of contributing to clinical prevention, diagnosis, treatment and improvement in prognosis.
Collapse
Affiliation(s)
- Junhao Hu
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, No.96 Fuxue Road, 325000, Wenzhou, People's Republic of China
| | - Wenyi Xu
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, No.96 Fuxue Road, 325000, Wenzhou, People's Republic of China
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, No.96 Fuxue Road, 325000, Wenzhou, People's Republic of China.
| | - Liangshan Mu
- School of Medicine, Zhejiang University, No.866 Yuhantang Road, 310058, Hangzhou, People's Republic of China.
| |
Collapse
|
9
|
Yaribeygi H, Maleki M, Sathyapalan T, Jamialahmadi T, Sahebkar A. Obesity and Insulin Resistance: A Review of Molecular Interactions. Curr Mol Med 2021; 21:182-193. [PMID: 32787760 DOI: 10.2174/1566524020666200812221527] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 07/05/2020] [Accepted: 07/12/2020] [Indexed: 12/06/2022]
Abstract
The prevalence of insulin resistance and diabetes mellitus is rising globally in epidemic proportions. Diabetes and its complications contribute to significant morbidity and mortality. An increase in sedentary lifestyle and consumption of a more energydense diet increased the incidence of obesity which is a significant risk factor for type 2 diabetes. Obesity acts as a potent upstream event that promotes molecular mechanisms involved in insulin resistance and diabetes mellitus. However, the exact molecular mechanisms between obesity and diabetes are not clearly understood. In the current study, we have reviewed the molecular interactions between obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Mina Maleki
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
| | | |
Collapse
|
10
|
KŠiŇanovÁ M, KovaŘÍkovÁ V, ŠefČÍkovÁ Z, ŠpirkovÁ A, ČikoŠ Š, Pisko J, Fabian D. Different response of embryos originating from control and obese mice to insulin in vitro. J Reprod Dev 2021; 67:25-34. [PMID: 33250503 PMCID: PMC7902211 DOI: 10.1262/jrd.2020-096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
The aim of the present work was to investigate the impact of maternal obesity on DNA methylation in ovulated oocytes, and to compare the response of in vitro-developing preimplantation embryos originating from control and obese mice to insulin. An intergenerational, diet-induced obesity model was used to produce outbred mice with an increased body weight and body fat. Two-cell and eight-cell embryos recovered from obese and control mice were cultured in a medium supplemented with 1 or 10 ng/ml insulin until blastocyst formation. In the derived blastocysts, cell proliferation, differentiation, and death rates were determined. The results of immunochemical visualization of 5-methylcytosine indicated a slightly higher DNA methylation in ovulated metaphase II oocytes recovered from obese females; however, the difference between groups did not reach statistical significance. Expanded blastocysts developed from embryos provided by control dams showed increased mean cell numbers (two and eight-cell embryos exposed to 10 ng/ml), an increased inner-cell-mass/trophectoderm ratio (two-cell embryos exposed to 1 ng/ml and eight-cell embryos exposed to 10 ng/ml), and a reduced level of apoptosis (two and eight-cell embryos exposed to 10 ng/ml). In contrast, embryos originating from obese mice were significantly less sensitive to insulin; indeed, no difference was recorded in any tested variable between the embryos exposed to insulin and those cultured in insulin-free medium. Real-time RT-PCR analysis showed a significant increase in the amount of insulin receptor transcripts in blastocysts recovered from obese dams. These results suggest that maternal obesity might modulate the mitogenic and antiapoptotic responses of preimplantation embryos to insulin.
Collapse
Affiliation(s)
- Martina KŠiŇanovÁ
- Institute of Animal Physiology, Centre of Biosciences, Slovak Academy of Sciences, 040 01 Košice, Slovak Republic
| | - Veronika KovaŘÍkovÁ
- Institute of Animal Physiology, Centre of Biosciences, Slovak Academy of Sciences, 040 01 Košice, Slovak Republic
| | - Zuzana ŠefČÍkovÁ
- Institute of Animal Physiology, Centre of Biosciences, Slovak Academy of Sciences, 040 01 Košice, Slovak Republic
| | - Alexandra ŠpirkovÁ
- Institute of Animal Physiology, Centre of Biosciences, Slovak Academy of Sciences, 040 01 Košice, Slovak Republic
| | - Štefan ČikoŠ
- Institute of Animal Physiology, Centre of Biosciences, Slovak Academy of Sciences, 040 01 Košice, Slovak Republic
| | - Jozef Pisko
- Institute of Animal Physiology, Centre of Biosciences, Slovak Academy of Sciences, 040 01 Košice, Slovak Republic
| | - Dušan Fabian
- Institute of Animal Physiology, Centre of Biosciences, Slovak Academy of Sciences, 040 01 Košice, Slovak Republic
| |
Collapse
|
11
|
Stafeev I, Sklyanik I, Mamontova E, Michurina S, Shestakova E, Yah’yaev K, Yurasov A, Masnikov D, Sineokaya M, Ratner E, Vorotnikov A, Menshikov M, Parfyonova Y, Shestakova M. NDRG1 Activity in Fat Depots Is Associated With Type 2 Diabetes and Impaired Incretin Profile in Patients With Morbid Obesity. Front Endocrinol (Lausanne) 2021; 12:777589. [PMID: 34956089 PMCID: PMC8695674 DOI: 10.3389/fendo.2021.777589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE We aimed to investigate insulin-, mTOR- and SGK1-dependent signaling basal states in morbidly obese patients' fat. We analyzed the correlation between the signaling activity, carbohydrate metabolism, and incretin profiles of patients. METHODS The omental and subcutaneous fat was obtained in patients with obesity. The omental study included 16 patients with normal glucose tolerance (NGT) and 17 patients with type 2 diabetes mellitus (T2DM); the subcutaneous study included 9 NGT patients and 12 T2DM patients. Insulin resistance was evaluated using the hyperinsulinemic euglycemic clamp test and HOMA-IR index. The oral glucose tolerance test (OGTT) for NGT patients and mixed meal tolerance test (MMTT) for T2DM patients were performed. The levels of incretins (GLP-1, GIP, oxyntomodulin) and glucagon were measured during the tests. Signaling was analyzed by Western blotting in adipose tissue biopsies. RESULTS We have shown equal levels of basal phosphorylation of insulin- and mTOR-dependent signaling in omental fat depot in NGT and T2DM obese patients. Nevertheless, pNDRG1-T346 was decreased in omental fat of T2DM patients. Correlation analysis has shown an inverse correlation of pNDRG1-T346 in omental fat and diabetic phenotype (HbA1c, impaired incretin profile (AUC GLP-1, glucagon)). Moreover, pNDRG1-T346 in subcutaneous fat correlated with impaired incretin levels among obese patients (inverse correlation with AUC glucagon and AUC GIP). CONCLUSIONS According to results of the present study, we hypothesize that phosphorylation of pNDRG1-T346 can be related to impairment in incretin hormone processing. pNDRG1-T346 in adipose tissue may serve as a marker of diabetes-associated impairments of the systemic incretin profile and insulin sensitivity.
Collapse
Affiliation(s)
- Iurii Stafeev
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
- *Correspondence: Iurii Stafeev,
| | - Igor Sklyanik
- Diabetes Institute, Endocrinology Research Centre, Moscow, Russia
| | - Elizaveta Mamontova
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
- Diabetes Institute, Endocrinology Research Centre, Moscow, Russia
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Svetlana Michurina
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
- Diabetes Institute, Endocrinology Research Centre, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | | | - Kamil Yah’yaev
- Surgery Department, Central Clinical Hospital #1 of Open Join Stock Company (OJSC) Russian Railways, Moscow, Russia
| | - Anatoliy Yurasov
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, Russia
- Surgery Department, Central Clinical Hospital #1 of Open Join Stock Company (OJSC) Russian Railways, Moscow, Russia
| | - Denis Masnikov
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
- Center of Master’s Programs, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Maria Sineokaya
- Diabetes Institute, Endocrinology Research Centre, Moscow, Russia
| | - Elizaveta Ratner
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
- Diabetes Institute, Endocrinology Research Centre, Moscow, Russia
| | - Alexander Vorotnikov
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
| | - Mikhail Menshikov
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
| | - Yelena Parfyonova
- The Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Moscow, Russia
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, Russia
| | | |
Collapse
|
12
|
Rawal K, Patel TP, Purohit KM, Israni K, Kataria V, Bhatt H, Gupta S. Influence of obese phenotype on metabolic profile, inflammatory mediators and stemness of hADSC in adipose tissue. Clin Nutr 2020; 39:3829-3835. [DOI: 10.1016/j.clnu.2020.02.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 11/25/2022]
|
13
|
Scalia P, Giordano A, Martini C, Williams SJ. Isoform- and Paralog-Switching in IR-Signaling: When Diabetes Opens the Gates to Cancer. Biomolecules 2020; 10:biom10121617. [PMID: 33266015 PMCID: PMC7761347 DOI: 10.3390/biom10121617] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Insulin receptor (IR) and IR-related signaling defects have been shown to trigger insulin-resistance in insulin-dependent cells and ultimately to give rise to type 2 diabetes in mammalian organisms. IR expression is ubiquitous in mammalian tissues, and its over-expression is also a common finding in cancerous cells. This latter finding has been shown to associate with both a relative and absolute increase in IR isoform-A (IR-A) expression, missing 12 aa in its EC subunit corresponding to exon 11. Since IR-A is a high-affinity transducer of Insulin-like Growth Factor-II (IGF-II) signals, a growth factor is often secreted by cancer cells; such event offers a direct molecular link between IR-A/IR-B increased ratio in insulin resistance states (obesity and type 2 diabetes) and the malignant advantage provided by IGF-II to solid tumors. Nonetheless, recent findings on the biological role of isoforms for cellular signaling components suggest that the preferential expression of IR isoform-A may be part of a wider contextual isoform-expression switch in downstream regulatory factors, potentially enhancing IR-dependent oncogenic effects. The present review focuses on the role of isoform- and paralog-dependent variability in the IR and downstream cellular components playing a potential role in the modulation of the IR-A signaling related to the changes induced by insulin-resistance-linked conditions as well as to their relationship with the benign versus malignant transition in underlying solid tumors.
Collapse
Affiliation(s)
- Pierluigi Scalia
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA; (A.G.); (C.M.); (S.J.W.)
- ISOPROG-Somatolink EPFP Network, Functional Research Unit, Philadelphia, PA 19104, USA and 93100 Caltanissetta, Italy
- Correspondence:
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA; (A.G.); (C.M.); (S.J.W.)
- Department of Medical Biotechnologies, University of Siena, 52100 Siena, Italy
| | - Caroline Martini
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA; (A.G.); (C.M.); (S.J.W.)
| | - Stephen J. Williams
- Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA 19122, USA; (A.G.); (C.M.); (S.J.W.)
- ISOPROG-Somatolink EPFP Network, Functional Research Unit, Philadelphia, PA 19104, USA and 93100 Caltanissetta, Italy
| |
Collapse
|
14
|
Hui C, Tomilov A, Garcia C, Jiang X, Fash DM, Khdour OM, Rosso C, Filippini G, Prato M, Graham J, Hecht S, Havel P, Cortopassi G. Novel idebenone analogs block Shc's access to insulin receptor to improve insulin sensitivity. Biomed Pharmacother 2020; 132:110823. [PMID: 33045613 DOI: 10.1016/j.biopha.2020.110823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/26/2020] [Accepted: 09/26/2020] [Indexed: 10/23/2022] Open
Abstract
There has been little innovation in identifying novel insulin sensitizers. Metformin, developed in the 1920s, is still used first for most Type 2 diabetes patients. Mice with genetic reduction of p52Shc protein have improved insulin sensitivity and glucose tolerance. By high-throughput screening, idebenone was isolated as the first small molecule 'Shc Blocker'. Idebenone blocks p52Shc's access to Insulin Receptor to increase insulin sensitivity. In this work the avidity of 34 novel idebenone analogs and 3 metabolites to bind p52Shc, and to block the interaction of p52Shc with the Insulin receptor was tested. Our hypothesis was that if an idebenone analog bound and blocked p52Shc's access to insulin receptor better than idebenone, it should be a more effective insulin sensitizing agent than idebenone itself. Of 34 analogs tested, only 2 both bound p52Shc more tightly and/or blocked the p52Shc-Insulin Receptor interaction more effectively than idebenone. Of those 2 only idebenone analog #11 was a superior insulin sensitizer to idebenone. Also, the long-lasting insulin-sensitizing potency of idebenone in rodents over many hours had been puzzling, as the parent molecule degrades to metabolites within 1 h. We observed that two of the idebenone's three metabolites are insulin sensitizing almost as potently as idebenone itself, explaining the persistent insulin sensitization of this rapidly metabolized molecule. These results help to identify key SAR = structure-activity relationship requirements for more potent small molecule Shc inhibitors as Shc-targeted insulin sensitizers for type 2 diabetes.
Collapse
Affiliation(s)
- ChunKiu Hui
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Alexey Tomilov
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Chase Garcia
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - XiaoSong Jiang
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - David M Fash
- Center for BioEnergetics, Biodesign Institute, Arizona State University, 1001 S McAllister Ave, Tempe, AZ, 85287, USA.
| | - Omar M Khdour
- Center for BioEnergetics, Biodesign Institute, Arizona State University, 1001 S McAllister Ave, Tempe, AZ, 85287, USA.
| | - Cristian Rosso
- Department of Chemical and Pharmaceutical Sciences, CENMAT, Center of Excellence for Nanostructured Materials, INSTM UdR, Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy.
| | - Giacomo Filippini
- Department of Chemical and Pharmaceutical Sciences, CENMAT, Center of Excellence for Nanostructured Materials, INSTM UdR, Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy.
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, CENMAT, Center of Excellence for Nanostructured Materials, INSTM UdR, Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy; Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014, Donostia San Sebastián, Spain; Basque Fdn Sci, Ikerbasque, Bilbao, 48013, Spain.
| | - James Graham
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Sidney Hecht
- Center for BioEnergetics, Biodesign Institute, Arizona State University, 1001 S McAllister Ave, Tempe, AZ, 85287, USA; School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA.
| | - Peter Havel
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Gino Cortopassi
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| |
Collapse
|
15
|
Regulation of lymphatic function and injury by nitrosative stress in obese mice. Mol Metab 2020; 42:101081. [PMID: 32941994 PMCID: PMC7536739 DOI: 10.1016/j.molmet.2020.101081] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022] Open
Abstract
Objective Obesity results in lymphatic dysfunction, but the cellular mechanisms that mediate this effect remain largely unknown. Previous studies in obese mice have shown that inducible nitric oxide synthase-expressing (iNOS+) inflammatory cells accumulate around lymphatic vessels. In the current study, we therefore tested the hypothesis that increased expression of iNOS results in nitrosative stress and injury to the lymphatic endothelial cells (LECs). In addition, we tested the hypothesis that lymphatic injury, independent of obesity, can modulate glucose and lipid metabolism. Methods We compared the metabolic changes and lymphatic function of wild-type and iNOS knockout mice fed a normal chow or high-fat diet for 16 weeks. To corroborate our in vivo findings, we analyzed the effects of reactive nitrogen species on isolated LECs. Finally, using a genetically engineered mouse model that allows partial ablation of the lymphatic system, we studied the effects of acute lymphatic injury on glucose and lipid metabolism in lean mice. Results The mesenteric lymphatic vessels of obese wild-type animals were dilated, leaky, and surrounded by iNOS+ inflammatory cells with resulting increased accumulation of reactive nitrogen species when compared with lean wild-type or obese iNOS knockout animals. These changes in obese wild-type mice were associated with systemic glucose and lipid abnormalities, as well as decreased mesenteric LEC expression of lymphatic-specific genes, including vascular endothelial growth factor receptor 3 (VEGFR-3) and antioxidant genes as compared with lean wild-type or obese iNOS knockout animals. In vitro experiments demonstrated that isolated LECs were more sensitive to reactive nitrogen species than blood endothelial cells, and that this sensitivity was ameliorated by antioxidant therapies. Finally, using mice in which the lymphatics were specifically ablated using diphtheria toxin, we found that the interaction between metabolic abnormalities caused by obesity and lymphatic dysfunction is bidirectional. Targeted partial ablation of mesenteric lymphatic channels of lean mice resulted in increased accumulation of iNOS+ inflammatory cells and increased reactive nitrogen species. Lymphatic ablation also caused marked abnormalities in insulin sensitivity, serum glucose and insulin concentrations, expression of insulin-sensitive genes, lipid metabolism, and significantly increased systemic and mesenteric white adipose tissue (M-WAT) inflammatory responses. Conclusions Our studies suggest that increased iNOS production in obese animals plays a key role in regulating lymphatic injury by increasing nitrosative stress. In addition, our studies suggest that obesity-induced lymphatic injury may amplify metabolic abnormalities by increasing systemic and local inflammatory responses and regulating insulin sensitivity. These findings suggest that manipulation of the lymphatic system may represent a novel means of treating metabolic abnormalities associated with obesity. Increased iNOS+ cells around mesenteric lymphatics of high fat diet-induced obese mice. iNOS knockout mice are protected from obesity-induced lymphatic dysfunction. Lymphatic endothelial cells are highly sensitive to nitrosative stress. Nitrosative stress causes lymphatic gene regulation. Lymphatic injury alone enhances iNOS+ cells and causes insulin resistance and dyslipidemia.
Collapse
|
16
|
Hosseini Khorami SA, Mutalib MSA, Feili Shiraz M, Abdullah JA, Rejali Z, Ali RM, Khaza'ai H. Genetic determinants of obesity heterogeneity in type II diabetes. Nutr Metab (Lond) 2020; 17:55. [PMID: 32670384 PMCID: PMC7346329 DOI: 10.1186/s12986-020-00476-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/01/2020] [Indexed: 11/23/2022] Open
Abstract
Background Although obesity is considered as the main cause of Type II diabetes (T2DM), non-obese individuals may still develop T2DM and obese individuals may not. Method The mRNA expression of PI3K/AKT axis from 100 non-obese and obese participants with insulin sensitivity and insulin resistance states were compared in this study toward the understanding of obesity heterogeneity molecular mechanism. Result In present study, there was no statistically significant difference in gene expression levels of IRS1 and PTEN between groups, whereas PI3K, AKT2 and GLUT4 genes were expressed at a lower level in obese diabetic group compared to other groups and were statistically significant. PDK1 gene was expressed at a higher level in non-obese diabetic group compared to obese diabetic and non-obese non-diabetics groups. No statistically significant difference was identified in gene expression pattern of PI3K/AKT pathway between obese non-diabetics and non-obese non-diabetics. Conclusion The components of PI3K/AKT pathway which is related to the fasting state, showed reduced expression in obese diabetic group due to the chronic over-nutrition which may induced insensitivity and reduced gene expression. The pathogenesis of insulin resistance in the absence of obesity in non-obese diabetic group could be due to disturbance in another pathway related to the non-fasting state like gluconeogenesis. Therefore, the molecular mechanism of insulin signalling in non-obese diabetic individuals is different from obese diabetics which more investigations are required to study insulin signalling pathways in greater depth, in order to assess nutritional factors, contribute to insulin resistance in obese diabetic and non-obese diabetic individuals.
Collapse
Affiliation(s)
| | - Mohd Sokhini Abd Mutalib
- Department of Nutrition and Dietetic, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Mohammad Feili Shiraz
- Department of Artificial Intelligence and Computer Engineering, Faculty of Electrical Engineering, Computer and IT, Qazvin Branch, Islamic Azad University, Qazvin, Iran
| | | | - Zulida Rejali
- Department of Obstetrics and Gynaecology, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Razana Mohd Ali
- Department of Pathology, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Huzwah Khaza'ai
- Department of Biomedical Science, University Putra Malaysia, 43400 Serdang, Selangor Malaysia
| |
Collapse
|
17
|
Abstract
Accumulating evidence highlights the importance of interactions between tumour cells and stromal cells for tumour initiation, progression, and metastasis. In tumours that contain adipocyte in their stroma, adipocytes contribute to modification of tumour microenvironment and affect metabolism of tumour and tumour progression by production of cytokines and adipokines from the lipids. The omentum and bone marrow (BM) are highly adipocyte-rich and are also common metastatic and primary tumour developmental sites. Omental adipocytes exhibit metabolic cross-talk, immune modulation, and angiogenesis. BM adipocytes secrete adipokines, and participate in solid tumour metastasis through regulation of the CCL2/CCR2 axis and metabolic interactions. BM adipocytes also contribute to the progression of hematopoietic neoplasms. Here, we here provide an overview of research progress on the cross-talks between omental/BM adipocytes and tumour cells, which may be pivotal modulators of tumour biology, thus highlighting novel therapeutic targets. Abbreviations: MCP-1, monocyte chemoattractant protein 1IL, interleukinSTAT3, signal transducer and activator of transcription 3FABP4, fatty acid binding protein 4PI3K/AKT, phosphoinositide 3-kinase/protein kinase BPPAR, peroxisome proliferator-activated receptorPUFA, polyunsaturated fatty acidTAM, tumour-associated macrophagesVEGF, vascular endothelial growth factorVEGFR, vascular endothelial growth factor receptorBM, bone marrowBMA, bone marrow adipocytesrBMA, regulated BMAcBMA, constitutive BMAUCP-1, uncoupling protein-1TNF-α, tumour necrosis factor-alphaRANKL, receptor activator of nuclear factor kappa-Β ligandVCAM-1, vascular cell adhesion molecule 1JAK2, Janus kinase 2CXCL (C–X–C motif) ligandPGE2, prostaglandin E2COX-2, cyclooxygenase-2CCL2, C-C motif chemokine ligand 2NF-κB, nuclear factor-kappa BMM, multiple myelomaALL, acute lymphoblastic leukemiaAML, acute myeloid leukemiaGDF15, growth differentiation factor 15AMPK, AMP-activated protein kinaseMAPK, mitogen-activated protein kinaseAPL, acute promyelocytic leukemiaCCR2, C-C motif chemokine receptor 2SDF-1α, stromal cell-derived factor-1 alphaFFA, free fatty acidsLPrA, leptin peptide receptor antagonistMCD, malonyl-CoA decarboxylase.
Collapse
Affiliation(s)
- Yoon Jin Cha
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Ja Seung Koo
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
18
|
Palachai N, Wattanathorn J, Muchimapura S, Thukham-mee W. Antimetabolic Syndrome Effect of Phytosome Containing the Combined Extracts of Mulberry and Ginger in an Animal Model of Metabolic Syndrome. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5972575. [PMID: 31827683 PMCID: PMC6881582 DOI: 10.1155/2019/5972575] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/16/2019] [Accepted: 09/21/2019] [Indexed: 12/15/2022]
Abstract
Due to the antimetabolic syndrome effect of mulberry and ginger together with the advantages of the synergistic effect and phytosome encapsulation technique, we hypothesized that phytosome containing the combined extracts of mulberry and ginger (PMG) should be able to manage MetS. PMG was developed and assessed the phenolic content and biological activities associated with the pathophysiology of MetS. The antimetabolic syndrome effect and the possible underlying mechanisms in the animal model of MetS were also assessed. Male Wistar rats induced MetS by subjecting to a 16-week high-carbohydrate high-fat diet. MetS rats were orally given PMG at doses of 50, 100, and 200 mg/kg for 21 days. They were determined metabolic parameter changes in serum, histomorphology changes of adipose tissue, the inflammatory cytokines such as IL-6 and TNF-α, oxidative stress status, PPAR-γ, and HDAC3 in adipose tissue. Our in vitro data showed that PMG increased phenolic contents and biological activities. PMG significantly improved MetS parameters including body weight gain, lipid profiles, plasma glucose, HOMA-IR, and ACE. In addition, the density and size of adipocyte, adiposity index, and weights of adipose tissues were also improved. Moreover, the decrease in TNF-α and IL-6, oxidative stress status, and HDAC3 expression together with the increase in PPAR-γ expression in adipose tissue was also observed. These data suggest that PMG exhibit antimetabolic syndrome and the possible underlying mechanism may be associated partly with the modulation effect on HDAC3, PPAR-γ, and adipose tissue. In addition, PMG also improves oxidative stress and inflammation in MetS. Therefore, PMG can be served as the potential supplement to manage MetS. However, a clinical trial study is essential to confirm this health benefit.
Collapse
Affiliation(s)
- Nut Palachai
- Department of Physiology and Graduate School (Neuroscience Program), Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
- Integrative Complementary Alternative Medicine Research, Khon Kaen University, Khon Kaen 40002, Thailand
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Jintanaporn Wattanathorn
- Integrative Complementary Alternative Medicine Research, Khon Kaen University, Khon Kaen 40002, Thailand
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Supaporn Muchimapura
- Integrative Complementary Alternative Medicine Research, Khon Kaen University, Khon Kaen 40002, Thailand
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wipawee Thukham-mee
- Integrative Complementary Alternative Medicine Research, Khon Kaen University, Khon Kaen 40002, Thailand
- Research Institute for Human High Performance and Health Promotion, Khon Kaen University, Khon Kaen 40002, Thailand
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
19
|
Withaferin A inhibits adipogenesis in 3T3-F442A cell line, improves insulin sensitivity and promotes weight loss in high fat diet-induced obese mice. PLoS One 2019; 14:e0218792. [PMID: 31226166 PMCID: PMC6588247 DOI: 10.1371/journal.pone.0218792] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 06/09/2019] [Indexed: 12/12/2022] Open
Abstract
The increased prevalence of obesity and associated insulin resistance calls for effective therapeutic treatment of metabolic diseases. The current PPARγ-targeting antidiabetic drugs have undesirable side effects. The present study investigated the anti-diabetic and anti-obesity effects of withaferin A (WFA) in diet-induced obese (DIO) C57BL/6J mice and also the anti-adipogenic effect of WFA in differentiating 3T3- F442A cells. DIO mice were treated with WFA (6 mg/kg) or rosiglitazone (10 mg/kg) for 8 weeks. At the end of the treatment period, metabolic profile, liver function and inflammatory parameters were obtained. Expression of selective genes controlling insulin signaling, inflammation, adipogenesis, energy expenditure and PPARγ phosphorylation-regulated genes in epididymal fats were analyzed. Furthermore, the anti-adipogenic effect of WFA was evaluated in 3T3- F442A cell line. WFA treatment prevented weight gain without affecting food or caloric intake in DIO mice. WFA-treated group also exhibited lower epididymal and mesenteric fat pad mass, an improvement in lipid profile and hepatic steatosis and a reduction in serum inflammatory cytokines. Insulin resistance was reduced as shown by an improvement in glucose and insulin tolerance and serum adiponectin. WFA treatment upregulated selective insulin signaling (insr, irs1, slc2a4 and pi3k) and PPARγ phosphorylation-regulated (car3, selenbp1, aplp2, txnip, and adipoq) genes, downregulated inflammatory (tnf-α and il-6) genes and altered energy expenditure controlling (tph2 and adrb3) genes. In 3T3- F442A cell line, withaferin A inhibited adipogenesis as indicated by a decrease in lipid accumulation in differentiating adipocytes and protein expression of PPARγ and C/EBPα. The effect of rosiglitazone on physiological and lipid profiles, insulin resistance, some genes expression and differentiating adipocytes were markedly different. Our data suggest that WFA is a promising therapeutic agent for both diabetes and obesity.
Collapse
|
20
|
Tomilov A, Allen S, Hui CK, Bettaieb A, Cortopassi G. Idebenone is a cytoprotective insulin sensitizer whose mechanism is Shc inhibition. Pharmacol Res 2018; 137:89-103. [PMID: 30290222 DOI: 10.1016/j.phrs.2018.09.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023]
Abstract
When insulin binds insulin receptor, IRS1 signaling is stimulated to trigger the maximal insulin response. p52Shc protein competes directly with IRS1, thus damping and diverting maximal insulin response. Genetic reduction of p52Shc minimizes competition with IRS1, and improves insulin signaling and glucose control in mice, and improves pathophysiological consequences of hyperglycemia. Given the multiple benefits of Shc reduction in vivo, we investigated whether any of 1680 drugs used in humans may function as Shc inhibitors, and thus potentially serve as novel anti-diabetics. Of the 1680, 30 insulin sensitizers were identified by screening in vitro, and of these 30 we demonstrated that 7 bound Shc protein. Of the 7 drugs, idebenone dose-dependently bound Shc protein in the 50-100 nM range, and induced insulin sensitivity and cytoprotection in this same 100 nM range that clinically dosed idebenone reaches in human plasma. By contrast we observe mitochondrial effects of idebenone in the 5,000 nM range that are not reached in human dosing. Multiple assays of target engagement demonstrate that idebenone physically interacts with Shc protein. Idebenone sensitizes mice to insulin in two different mouse models of prediabetes. Genetic depletion of idebenone's target eliminates idebenone's ability to insulin-sensitize in vivo. Thus, idebenone is the first-in-class member of a novel category of insulin-sensitizing and cytoprotective agents, the Shc inhibitors. Idebenone is an approved drug and could be considered for other indications such as type 2 diabetes and fatty liver disease, in which insulin resistance occurs.
Collapse
Affiliation(s)
- Alexey Tomilov
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Sonia Allen
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Chun Kiu Hui
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Ahmed Bettaieb
- Department of Nutrition, The University of Tennessee, 1215 W. Cumberland Ave, Knoxville, TN, 37996-1920, USA.
| | - Gino Cortopassi
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| |
Collapse
|
21
|
Timpson AJ, de Mestre AM, Elliott J, Harris PA, Cheng Z, Mirczuk SM, Callan L, Rainbow L, Menzies-Gow NJ. Seasonal and Dietary Influences on Adipose Tissue and Systemic Gene Expression in Control and Previously Laminitic Ponies. J Equine Vet Sci 2018. [DOI: 10.1016/j.jevs.2018.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
22
|
Jung J, Kim GW, Lee W, Mok C, Chung SH, Jang W. Meta- and cross-species analyses of insulin resistance based on gene expression datasets in human white adipose tissues. Sci Rep 2018; 8:3747. [PMID: 29487289 PMCID: PMC5829071 DOI: 10.1038/s41598-017-18082-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 12/06/2017] [Indexed: 01/08/2023] Open
Abstract
Ample evidence indicates that insulin resistance (IR) is closely related to white adipose tissue (WAT), but the underlying mechanisms of IR pathogenesis are still unclear. Using 352 microarray datasets from seven independent studies, we identified a meta-signature which comprised of 1,413 genes. Our meta-signature was also enriched in overall WAT in in vitro and in vivo IR models. Only 12 core enrichment genes were consistently enriched across all IR models. Among the meta-signature, we identified a drug signature made up of 211 genes with expression levels that were co-regulated by thiazolidinediones and metformin using cross-species analysis. To confirm the clinical relevance of our drug signature, we found that the expression levels of 195 genes in the drug signature were significantly correlated with both homeostasis model assessment 2-IR score and body mass index. Finally, 18 genes from the drug signature were identified by protein-protein interaction network cluster. Four core enrichment genes were included in 18 genes and the expression levels of selected 8 genes were validated by quantitative PCR. These findings suggest that our signatures provide a robust set of genetic markers which can be used to provide a starting point for developing potential therapeutic targets in improving IR in WAT.
Collapse
Affiliation(s)
- Junghyun Jung
- Department of Life Science, Dongguk University, 30 Pildong ro 1-gil, 04620, Seoul, Korea
| | - Go Woon Kim
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, 02447, Seoul, Korea
| | - Woosuk Lee
- Department of Life Science, Dongguk University, 30 Pildong ro 1-gil, 04620, Seoul, Korea
| | - Changsoo Mok
- Department of Life Science, Dongguk University, 30 Pildong ro 1-gil, 04620, Seoul, Korea
| | - Sung Hyun Chung
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, 02447, Seoul, Korea
| | - Wonhee Jang
- Department of Life Science, Dongguk University, 30 Pildong ro 1-gil, 04620, Seoul, Korea.
| |
Collapse
|
23
|
Khawaja KI, Mian SA, Fatima A, Tahir GM, Khan FF, Burney S, Hasan A, Masud F. Phenotypic and metabolic dichotomy in obesity: clinical, biochemical and immunological correlates of metabolically divergent obese phenotypes in healthy South Asian adults. Singapore Med J 2018; 59:431-438. [PMID: 29430577 DOI: 10.11622/smedj.2018019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Metabolic heterogeneity among obese individuals is thought to translate into variations in cardiovascular risk. Identifying obese people with an unfavourable metabolic profile may allow preventive strategies to be targeted at high-risk groups. This study aimed to identify clinical, biochemical and immunological differences between insulin-sensitive and insulin-resistant obese subgroups, to understand the population-specific pathophysiological basis of the adverse cardiovascular risk profile in the latter group. METHODS Cardiovascular risk indicators, including anthropometric parameters, blood pressure, acanthosis nigricans area, and related biochemical, endocrine and inflammatory markers, were determined in 255 healthy South Asian volunteers aged 18-45 years, with a 2:1 ratio of obese/overweight to normal-weight individuals. Lifetime atherosclerotic cardiovascular disease (ASCVD) risk was also calculated. RESULTS Body mass index (BMI) and insulin sensitivity-based tertiles independently showed incremental trends in waist-hip ratio, skinfold thickness, acanthosis nigricans area, blood pressure, serum lipids, hepatic enzymes, adipokines, inflammatory markers and ten-year ASCVD risk. The anthropometric, biochemical and inflammatory parameters of obese insulin-sensitive and obese insulin-resistant groups differed significantly. Extreme group analysis after excluding the middle tertiles of both insulin resistance and BMI also showed significant difference in anthropometric indicators of cardiovascular risk and estimated lifetime ASCVD risk between the two obese subgroups. CONCLUSION Obese insulin-sensitive individuals had a favourable metabolic profile compared to the obese insulin-resistant group. The most consistent discriminative factor between these phenotypic classes was anthropometric parameters, which underscores the importance of clinical parameters as cardiovascular risk indicators in obesity.
Collapse
Affiliation(s)
- Khadija Irfan Khawaja
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences and Services Hospital, Lahore, Pakistan
| | - Saqib Ali Mian
- Diabetes Care Centre, King Salman Hospital, Riyadh, Kingdom of Saudi Arabia
| | - Aziz Fatima
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences and Services Hospital, Lahore, Pakistan
| | - Ghulam Murtaza Tahir
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences and Services Hospital, Lahore, Pakistan
| | - Fehmida Farrukh Khan
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences and Services Hospital, Lahore, Pakistan
| | - Saira Burney
- Department of Endocrinology and Metabolism, Services Institute of Medical Sciences and Services Hospital, Lahore, Pakistan
| | - Ali Hasan
- Medical Unit No. 4, Services Institute of Medical Sciences and Services Hospital, Lahore, Pakistan
| | - Faisal Masud
- King Edward Medical University, Lahore, Pakistan
| |
Collapse
|
24
|
Yan K, Zhu H, Xu J, Pan H, Li N, Wang L, Yang H, Liu M, Gong F. Lotus Leaf Aqueous Extract Reduces Visceral Fat Mass and Ameliorates Insulin Resistance in HFD-Induced Obese Rats by Regulating PPARγ2 Expression. Front Pharmacol 2017; 8:409. [PMID: 28690544 PMCID: PMC5481353 DOI: 10.3389/fphar.2017.00409] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/09/2017] [Indexed: 12/18/2022] Open
Abstract
Objectives: Lotus leaf is a kind of traditional Chinese medicine. We aimed to explore the effects of lotus leaf aqueous extract (LLAE) on peroxisome proliferative activated receptor γ2 (PPARγ2) expression in preadipocytes and adipocytes and further investigate its effects on high fat diet (HFD)-induced obese rats. Methods: pGL3-Enhancer-PPARγ2 (625 bp)-Luc plasmid, a luciferase reporter gene expression plasmid containing PPARγ2 promoter, was stably transfected into 3T3-L1 preadipocytes. PPARγ2 promoter activities were determined by assaying the luciferase activities. Then PPARγ2 promoter activities in preadipocytes and PPARγ2 mRNA levels in human subcutaneous adipocytes were measured after the administration with LLAE. Additionally, the effects of LLAE on body weight, fat mass, glucose and lipid metabolism and the expression of PPARγ2, insulin receptor substrate 1 and glucose transporter 4 (GLUT4) in visceral adipose tissue (VAT) were measured in HFD-induced obese rats treated with low or high dose [0.5 or 3.0 g crude drug/(kg.d)] LLAE for 6 weeks. Results: Ten μg/ml LLAE significantly increased the luciferase activities in 3T3-L1 cells and the stimulatory action reached 2.51 folds of controls when LLAE was 1000 μg/ml (P < 0.01). After treating 3T3-L1 cells with 100 μg/ml LLAE, the stimulatory role peaked at 32 h where it was 2.58 folds of controls (P < 0.01). Besides, 100 μg/ml LLAE significantly increased PPARγ2 mRNA levels in human adipocytes to 1.91 folds of controls (P < 0.01). In HFD-induced obese rats, administration with both low and high dose LLAE notably reduced visceral fat mass by 45.5 and 58.4%, respectively, and significantly decreased fasting serum insulin levels (P < 0.05). The high dose LLAE also significantly decreased homeostasis model assessment of insulin resistance in obese rats (P < 0.05). Furthermore, the mRNA levels of PPARγ2 and GLUT4 in VAT of obese rats were significantly increased when compared with control rats, and were notably suppressed by LLAE intervention for 6 weeks (P < 0.05). Conclusion: LLAE significantly reduces visceral fat mass and ameliorates insulin resistance in HFD-induced obese rats. These beneficial effects of LLAE may associate with its role in stimulating PPARγ2 expression in preadipocytes and subcutaneous adipocytes and suppressing PPARγ2 and GLUT4 expression in VAT.
Collapse
Affiliation(s)
- Kemin Yan
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Jian Xu
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China.,Department of Endocrinology, Beijing Tian Tan Hospital, Capital Medical UniversityBeijing, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Naishi Li
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - Meijuan Liu
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| | - FengYing Gong
- Key Laboratory of Endocrinology of National Health and Family Planning Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
25
|
Gonzalez-Franquesa A, Patti ME. Insulin Resistance and Mitochondrial Dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:465-520. [DOI: 10.1007/978-3-319-55330-6_25] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Anatomical distribution of primary amine oxidase activity in four adipose depots and plasma of severely obese women with or without a dysmetabolic profile. J Physiol Biochem 2016; 73:475-486. [PMID: 27766585 DOI: 10.1007/s13105-016-0526-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/29/2016] [Indexed: 12/21/2022]
Abstract
Semicarbazide-sensitive amine oxidase (SSAO), identical to primary amine oxidase or vascular adhesion protein-1, is a membrane enzyme that generates hydrogen peroxide. SSAO is highly expressed at the adipocyte surface, and its plasma levels increase with type 2 diabetes. Since visceral adipose tissue (AT) is more tightly associated with obesity complications than subcutaneous (SC) abdominal fat, we compared SSAO activity in plasma and 4 distinct AT locations in 48 severely obese women (body mass index (BMI), averaging 54 ± 11 kg/m2), with or without a dysmetabolic profile. Higher glucose and triacylglycerol levels vs lower high-density lipoprotein (HDL)-cholesterol characterized dysmetabolic women (DYS; n = 25) from non-dysmetabolic (NDYS; n = 23), age- and weight-matched subjects. SC, mesenteric (ME), omental (OM), and round ligament (RL) fat locations were collected during bariatric surgery. SSAO capacity to oxidize up to 1 mM benzylamine was determined in AT and plasma with radiometric and fluorimetric methods. Plasma SSAO was higher in the DYS group. SSAO activity was higher in fat than in plasma, when expressed as radiolabeled benzaldehyde per milligram of protein. In ATs from DYS women, protein content was 10 % higher, and basal hydrogen peroxide release lower than in NDYS subjects, except for RL location. The SSAO affinity towards benzylamine did not exhibit regional variation and was not altered by a dysmetabolic profile (K m averaging 184 ± 7 μM; n = 183). Although radiometric and fluorimetric methods gave different estimates of oxidase activity, both indicated that AT SSAO activity did not vary according to anatomical location and/or metabolic status in severely obese women.
Collapse
|
27
|
Mediterranean diet and mortality risk in metabolically healthy obese and metabolically unhealthy obese phenotypes. Int J Obes (Lond) 2016; 40:1541-1549. [DOI: 10.1038/ijo.2016.114] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 06/04/2016] [Accepted: 06/12/2016] [Indexed: 12/15/2022]
|
28
|
Moreno-Navarrete JM, Jove M, Ortega F, Xifra G, Ricart W, Obis È, Pamplona R, Portero-Otin M, Fernández-Real JM. Metabolomics uncovers the role of adipose tissue PDXK in adipogenesis and systemic insulin sensitivity. Diabetologia 2016; 59:822-32. [PMID: 26831303 DOI: 10.1007/s00125-016-3863-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/17/2015] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS We aimed to investigate the potential mechanisms involved in the compromised adipogenesis of visceral (VAT) vs subcutaneous adipose tissue (SAT) using comparative metabolomics. Based on the differentially identified metabolites, we focused on the relationship between the active form of vitamin B6 (pyridoxal 5-phosphate [PLP]), known to be generated through pyridoxal kinase (PDXK), and adipogenesis. METHODS Non-targeted metabolomics analyses were performed in paired VAT and SAT (n = 14, discovery cohort). PDXK gene expression was evaluated in two validation cohorts of paired SAT and VAT samples in relation to obesity status and insulin sensitivity, and mechanistically after weight loss in vivo and in 3T3-L1 cells in vitro. RESULTS Comparative metabolomics showed that PLP was significantly decreased in VAT vs SAT. Concordantly, PDXK mRNA levels were significantly decreased in VAT vs SAT, specifically in adipocytes. The decrease was specially marked in obese individuals. PDXK mRNA levels showed a strong association with adipogenic, lipid-droplet-related and lipogenic genes. At a functional level, systemic insulin sensitivity positively associated with PDXK expression, and surgically-induced weight loss (improving insulin sensitivity) led to increased SAT PDXK mRNA levels in parallel with adipogenic genes. In human pre-adipocytes, PDXK mRNA levels increased during adipocyte differentiation and after administration of peroxisome proliferator-activated receptor-γ agonists, and decreased under inflammatory stimuli. Mechanistic studies in 3T3-L1 cells showed that PLP administration resulted in increased adipogenic mRNA markers during early adipogenesis, whereas the PLP antagonist 4-deoxypyridoxine exerted opposite effects. CONCLUSIONS/INTERPRETATION Overall, these results support the notion that in situ production of PLP is required for physiological adipogenesis.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Mariona Jove
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (IRBLleida)-PCiTAL, Lleida, Spain
| | - Francisco Ortega
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Gemma Xifra
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Wifredo Ricart
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Èlia Obis
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (IRBLleida)-PCiTAL, Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (IRBLleida)-PCiTAL, Lleida, Spain
| | - Manuel Portero-Otin
- Department of Experimental Medicine, University of Lleida-Biomedical Research Institute of Lleida (IRBLleida)-PCiTAL, Lleida, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Hospital of Girona 'Dr Josep Trueta', Carretera de França s/n, 17007, Girona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain, .
| |
Collapse
|
29
|
Kirby TJ, Walton RG, Finlin B, Zhu B, Unal R, Rasouli N, Peterson CA, Kern PA. Integrative mRNA-microRNA analyses reveal novel interactions related to insulin sensitivity in human adipose tissue. Physiol Genomics 2016; 48:145-53. [PMID: 26672043 PMCID: PMC4729698 DOI: 10.1152/physiolgenomics.00071.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 12/11/2015] [Indexed: 01/17/2023] Open
Abstract
Adipose tissue has profound effects on whole-body insulin sensitivity. However, the underlying biological processes are quite complex and likely multifactorial. For instance, the adipose transcriptome is posttranscriptionally modulated by microRNAs, but the relationship between microRNAs and insulin sensitivity in humans remains to be determined. To this end, we utilized an integrative mRNA-microRNA microarray approach to identify putative molecular interactions that regulate the transcriptome in subcutaneous adipose tissue of insulin-sensitive (IS) and insulin-resistant (IR) individuals. Using the NanoString nCounter Human v1 microRNA Expression Assay, we show that 17 microRNAs are differentially expressed in IR vs. IS. Of these, 16 microRNAs (94%) are downregulated in IR vs. IS, including miR-26b, miR-30b, and miR-145. Using Agilent Human Whole Genome arrays, we identified genes that were predicted targets of miR-26b, miR-30b, and miR-145 and were upregulated in IR subjects. This analysis produced ADAM22, MYO5A, LOX, and GM2A as predicted gene targets of these microRNAs. We then validated that miR-145 and miR-30b regulate these mRNAs in differentiated human adipose stem cells. We suggest that use of bioinformatic integration of mRNA and microRNA arrays yields verifiable mRNA-microRNA pairs that are associated with insulin resistance and can be validated in vitro.
Collapse
Affiliation(s)
- Tyler J Kirby
- College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - R Grace Walton
- College of Health Sciences, University of Kentucky, Lexington, Kentucky
| | - Brian Finlin
- Department of Medicine, Division of Endocrinology, and Barnstable Brown Diabetes and Obesity Center; University of Kentucky, Lexington, Kentucky; and
| | - Beibei Zhu
- Department of Medicine, Division of Endocrinology, and Barnstable Brown Diabetes and Obesity Center; University of Kentucky, Lexington, Kentucky; and
| | - Resat Unal
- Department of Medicine, Division of Endocrinology, and Barnstable Brown Diabetes and Obesity Center; University of Kentucky, Lexington, Kentucky; and
| | - Neda Rasouli
- Department of Internal Medicine, Division of Endocrinology, University of Colorado, Aurora, Colorado
| | | | - Philip A Kern
- Department of Medicine, Division of Endocrinology, and Barnstable Brown Diabetes and Obesity Center; University of Kentucky, Lexington, Kentucky; and
| |
Collapse
|
30
|
Roles of Oxidative Stress in Polycystic Ovary Syndrome and Cancers. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:8589318. [PMID: 26770659 PMCID: PMC4684888 DOI: 10.1155/2016/8589318] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/28/2015] [Accepted: 09/06/2015] [Indexed: 12/16/2022]
Abstract
Oxidative stress (OS) has received extensive attention in the last two decades, because of the discovery that abnormal oxidation status was related to patients with chronic diseases, such as diabetes, cardiovascular, polycystic ovary syndrome (PCOS), cancer, and neurological diseases. OS is considered as a potential inducing factor in the pathogenesis of PCOS, which is one of the most common complex endocrine disorders and a leading cause of female infertility, affecting 4%–12% of women in the world, as OS has close interactions with PCOS characteristics, just as insulin resistance (IR), hyperandrogenemia, and chronic inflammation. It has also been shown that DNA mutations and alterations induced by OS are involved in cancer pathogenesis, tumor cell survival, proliferation, invasion, angiogenesis, and so on. Furthermore, recent studies show that the females with PCOS are reported to have an increasing risk of cancers. As a result, the more serious OS in PCOS is regarded as an important potential incentive for the increasing risk of cancers, and this study aims to analyze the possibility and potential pathogenic mechanism of the above process, providing insightful thoughts and evidences for preventing cancer potentially caused by PCOS in clinic.
Collapse
|
31
|
Gene expression of different adipose tissues of severely obese women with or without a dysmetabolic profile. J Physiol Biochem 2015; 71:719-32. [DOI: 10.1007/s13105-015-0436-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/14/2015] [Indexed: 01/04/2023]
|
32
|
Kato Y, Kamiya H, Koide N, Odkhuu E, Komatsu T, Dagvadorj J, Watarai A, Kondo M, Kato K, Nakamura J, Yokochi T. Spironolactone inhibits production of proinflammatory mediators in response to lipopolysaccharide via inactivation of nuclear factor-κB. Immunopharmacol Immunotoxicol 2015; 36:237-41. [PMID: 24852317 DOI: 10.3109/08923973.2014.921690] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The effect of spironolactone (SPIR) on lipopolysaccharide (LPS)-induced production of proinflammatory mediators was examined using RAW 264.7 macrophage-like cells and mouse peritoneal macrophages. SPIR significantly inhibited LPS-induced production of nitric oxide (NO), tumor necrosis factor-α and prostaglandin E2. The inhibition was not mediated by cell death. SPIR reduced the expression of an inducible NO synthase mRNA in response to LPS. SPIR significantly inhibited phosphorylation of p65 nuclear factor (NF)-κB in response to LPS. Furthermore, SPIR inhibited phosphorylation of IκB kinase (IKK) as an upstream molecule of NF-κB in response to LPS. LPS did not induce the production of aldosterone in RAW 264.7 cells. Taken together, SPIR is suggested to inhibit LPS-induced proinflammatory mediators via inactivation of IKK/NF-κB in LPS signaling.
Collapse
Affiliation(s)
- Yoshiro Kato
- Division of Diabetes, Department of Internal Medicine, Diabetes Center , Nagakute , Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kogelman LJA, Cirera S, Zhernakova DV, Fredholm M, Franke L, Kadarmideen HN. Identification of co-expression gene networks, regulatory genes and pathways for obesity based on adipose tissue RNA Sequencing in a porcine model. BMC Med Genomics 2014; 7:57. [PMID: 25270054 PMCID: PMC4183073 DOI: 10.1186/1755-8794-7-57] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/24/2014] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Obesity is a complex metabolic condition in strong association with various diseases, like type 2 diabetes, resulting in major public health and economic implications. Obesity is the result of environmental and genetic factors and their interactions, including genome-wide genetic interactions. Identification of co-expressed and regulatory genes in RNA extracted from relevant tissues representing lean and obese individuals provides an entry point for the identification of genes and pathways of importance to the development of obesity. The pig, an omnivorous animal, is an excellent model for human obesity, offering the possibility to study in-depth organ-level transcriptomic regulations of obesity, unfeasible in humans. Our aim was to reveal adipose tissue co-expression networks, pathways and transcriptional regulations of obesity using RNA Sequencing based systems biology approaches in a porcine model. METHODS We selected 36 animals for RNA Sequencing from a previously created F2 pig population representing three extreme groups based on their predicted genetic risks for obesity. We applied Weighted Gene Co-expression Network Analysis (WGCNA) to detect clusters of highly co-expressed genes (modules). Additionally, regulator genes were detected using Lemon-Tree algorithms. RESULTS WGCNA revealed five modules which were strongly correlated with at least one obesity-related phenotype (correlations ranging from -0.54 to 0.72, P < 0.001). Functional annotation identified pathways enlightening the association between obesity and other diseases, like osteoporosis (osteoclast differentiation, P = 1.4E-7), and immune-related complications (e.g. Natural killer cell mediated cytotoxity, P = 3.8E-5; B cell receptor signaling pathway, P = 7.2E-5). Lemon-Tree identified three potential regulator genes, using confident scores, for the WGCNA module which was associated with osteoclast differentiation: CCR1, MSR1 and SI1 (probability scores respectively 95.30, 62.28, and 34.58). Moreover, detection of differentially connected genes identified various genes previously identified to be associated with obesity in humans and rodents, e.g. CSF1R and MARC2. CONCLUSIONS To our knowledge, this is the first study to apply systems biology approaches using porcine adipose tissue RNA-Sequencing data in a genetically characterized porcine model for obesity. We revealed complex networks, pathways, candidate and regulatory genes related to obesity, confirming the complexity of obesity and its association with immune-related disorders and osteoporosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Haja N Kadarmideen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 7, 1870, Frederiksberg, Denmark.
| |
Collapse
|
34
|
Chen L, Xu WM, Zhang D. Association of abdominal obesity, insulin resistance, and oxidative stress in adipose tissue in women with polycystic ovary syndrome. Fertil Steril 2014; 102:1167-1174.e4. [PMID: 25064406 DOI: 10.1016/j.fertnstert.2014.06.027] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 05/25/2014] [Accepted: 06/17/2014] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To study the expression of insulin signaling-related genes and oxidative stress markers in the visceral adipose tissue obtained from polycystic ovary syndrome (PCOS) patients and healthy control subjects and to investigate the relationships among abdominal obesity, insulin resistance, and oxidative stress at the tissue level. DESIGN Case-control study. SETTING University teaching hospital. PATIENT(S) In total, 30 PCOS patients and 30 healthy control subjects, who underwent laparoscopic surgery, were included in the study. INTERVENTION(S) Abdominal obesity was defined based on waist circumference (WC). The homeostasis model index was used to assess insulin resistance (HOMA-IR). MAIN OUTCOME MEASURE(S) Gene expression of glucose transporter 4 (GLUT4) and insulin receptor substrate 1 (IRS1) in visceral adipose tissue (VAT) and the parameters of oxidative stress, such as superoxide dismutase, enzyme glutathione reductase, and dimethylarginine, were measured, and the expression of protein oxidative damage product 3-nitro-tyrosine residues (nitrotyrosine) in VAT was identified with the use of immunohistochemistry. RESULT(S) PCOS was associated with lower expression of GLUT4 and IRS1 and a higher level of oxidative stress in VAT, which was strongly correlated with WC and HOMA-IR. Presence of abdominal obesity further intensified the correlations observed in our measurements. The nitrotyrosine expression in VAT was stronger in PCOS patients. CONCLUSION(S) The strong correlation of insulin resistance with oxidative stress at the VAT level suggests that local oxidative stress and abnormalities of insulin signaling in adipose tissue play critical roles in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Li Chen
- Department of Obstetrics and Gynecology, Sichuan University-Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Wen Ming Xu
- Joint Laboratory for Reproductive Medicine, Sichuan University-Chinese University of Hong Kong, Hong Kong, People's Republic of China; Key Laboratory of Obstetric, Gynecologic, and Pediatric Diseases and Birth Defects, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Dan Zhang
- Department of Obstetrics and Gynecology, Sichuan University-Chinese University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
35
|
Expression and regulation of facilitative glucose transporters in equine insulin-sensitive tissue: from physiology to pathology. ISRN VETERINARY SCIENCE 2014; 2014:409547. [PMID: 24977043 PMCID: PMC4060548 DOI: 10.1155/2014/409547] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/09/2013] [Indexed: 01/12/2023]
Abstract
Glucose uptake is the rate-limiting step in glucose utilization in mammalians and is tightly regulated by a family of specialized proteins, called the facilitated glucose transporters (GLUTs/SLC2). GLUT4, the major isoform in insulin-responsive tissue, translocates from an intracellular pool to the cell surface and as such determines insulin-stimulated glucose uptake. However, despite intensive research over 50 years, the insulin-dependent and -independent pathways that mediate GLUT4 translocation are not fully elucidated in any species. Insulin resistance (IR) is one of the hallmarks of equine metabolic syndrome and is the most common metabolic predisposition for laminitis in horses. IR is characterized by the impaired ability of insulin to stimulate glucose disposal into insulin-sensitive tissues. Similar to other species, the functional capability of the insulin-responsive GLUTs is impaired in muscle and adipose tissue during IR in horses. However, the molecular mechanisms of altered glucose transport remain elusive in all species, and there is still much to learn about the physiological and pathophysiological functions of the GLUT family members, especially in regard to class III. Since GLUTs are key regulators of whole-body glucose homeostasis, they have received considerable attention as potential therapeutic targets to treat metabolic disorders in human and equine patients.
Collapse
|
36
|
Peral B, Camafeita E, Fernández-Real JM, López JA. Tackling the human adipose tissue proteome to gain insight into obesity and related pathologies. Expert Rev Proteomics 2014; 6:353-61. [DOI: 10.1586/epr.09.53] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
37
|
Gnanasegaran N, Govindasamy V, Musa S, Kasim NHA. Different isolation methods alter the gene expression profiling of adipose derived stem cells. Int J Med Sci 2014; 11:391-403. [PMID: 24669199 PMCID: PMC3964446 DOI: 10.7150/ijms.7697] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/20/2014] [Indexed: 12/22/2022] Open
Abstract
Human adipose stem cells (ASCs) has been in the limelight since its discovery as a suitable source of mesenchymal stem cells (MSCs) in regenerative medicine. Currently, two major techniques are used to isolate ASCs, namely liposuction and tissue biopsy. These two methods are relatively risk-free but the question as to which method could give a more efficient output remains unclear. Thus, this study was carried out to compare and contrast the output generated in regards to growth kinetics, differentiation capabilities in vitro, and gene expression profiling. It was found that ASCs from both isolation methods were comparable in terms of growth kinetics and tri-lineage differentiation. Furthermore, ASCs from both populations were reported as CD44(+), CD73(+), CD90(+), CD166(+), CD34(-), CD45(-) and HLA-DR(-). However, in regards to gene expression, a group of overlapping genes as well as distinct genes were observed. Distinct gene expressions indicated that ASCs (liposuction) has endoderm lineage propensity whereas ASCs (biopsy) has a tendency towards mesoderm/ectoderm lineage. This information suggests involvement in different functional activity in accordance to isolation method. In conclusion, future studies to better understand these gene functions should be carried out in order to contribute in the applicability of each respective cells in regenerative therapy.
Collapse
Affiliation(s)
- Nareshwaran Gnanasegaran
- 1. Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Vijayendran Govindasamy
- 2. Hygieia Innovation Sdn. Bhd, Lot 1G-2G, Lanai Complex No.2, Persiaran Seri Perdana, Persint 10, Federal Territory of Putrajaya, Malaysia
| | - Sabri Musa
- 1. Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia ; 3. Department of Children's Dentistry and Orthodontics, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- 1. Regenerative Dentistry Research Group (ReDReG), Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia ; 4. Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Jové M, Moreno-Navarrete JM, Pamplona R, Ricart W, Portero-Otín M, Fernández-Real JM. Human omental and subcutaneous adipose tissue exhibit specific lipidomic signatures. FASEB J 2013; 28:1071-81. [PMID: 24265485 DOI: 10.1096/fj.13-234419] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite their differential effects on human metabolic pathophysiology, the differences in omental and subcutaneous lipidomes are largely unknown. To explore this field, liquid chromatography coupled with mass spectrometry was used for lipidome analyses of adipose tissue samples (visceral and subcutaneous) selected from a group of obese subjects (n=38). Transcriptomics and in vitro studies in adipocytes were used to confirm the pathways affected by location. The analyses revealed the existence of obesity-related specific lipidome signatures in each of these locations, attributed to selective enrichment of specific triglycerides, glycerophospholipids, and sphingolipids, because these were not observed in adipose tissues from nonobese individuals. The changes were compatible with subcutaneous enrichment in pathways involved in adipogenesis, triacylglyceride synthesis, and lipid droplet formation, as well as increased α-oxidation. Marked differences between omental and subcutaneous depots in obese individuals were seen in the association of lipid species with metabolic traits (body mass index and insulin sensitivity). Targeted studies also revealed increased cholesterol (Δ56%) and cholesterol epoxide (Δ34%) concentrations in omental adipose tissue. In view of the effects of cholesterol epoxide, which induced enhanced expression of adipocyte differentiation and α-oxidation genes in human omental adipocytes, a novel role for cholesterol epoxide as a signaling molecule for differentiation is proposed. In summary, in obesity, adipose tissue exhibits a location-specific differential lipid profile that may contribute to explaining part of its distinct pathogenic role.
Collapse
Affiliation(s)
- Mariona Jové
- 1Section of Diabetes, Endocrinology and Nutrition, Hospital of Girona "Dr Josep Trueta," Carretera de França s/n, 17007, Girona, Spain.
| | | | | | | | | | | |
Collapse
|
39
|
Garcia-Arcos I, Hiyama Y, Drosatos K, Bharadwaj KG, Hu Y, Son NH, O'Byrne SM, Chang CL, Deckelbaum RJ, Takahashi M, Westerterp M, Obunike JC, Jiang H, Yagyu H, Blaner WS, Goldberg IJ. Adipose-specific lipoprotein lipase deficiency more profoundly affects brown than white fat biology. J Biol Chem 2013; 288:14046-14058. [PMID: 23542081 DOI: 10.1074/jbc.m113.469270] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adipose fat storage is thought to require uptake of circulating triglyceride (TG)-derived fatty acids via lipoprotein lipase (LpL). To determine how LpL affects the biology of adipose tissue, we created adipose-specific LpL knock-out (ATLO) mice, and we compared them with whole body LpL knock-out mice rescued with muscle LpL expression (MCK/L0) and wild type (WT) mice. ATLO LpL mRNA and activity were reduced, respectively, 75 and 70% in gonadal adipose tissue (GAT), 90 and 80% in subcutaneous tissue, and 84 and 85% in brown adipose tissue (BAT). ATLO mice had increased plasma TG levels associated with reduced chylomicron TG uptake into BAT and lung. ATLO BAT, but not GAT, had altered TG composition. GAT from MCK/L0 was smaller and contained less polyunsaturated fatty acids in TG, although GAT from ATLO was normal unless LpL was overexpressed in muscle. High fat diet feeding led to less adipose in MCK/L0 mice but TG acyl composition in subcutaneous tissue and BAT reverted to that of WT. Therefore, adipocyte LpL in BAT modulates plasma lipoprotein clearance, and the greater metabolic activity of this depot makes its lipid composition more dependent on LpL-mediated uptake. Loss of adipose LpL reduces fat accumulation only if accompanied by greater LpL activity in muscle. These data support the role of LpL as the "gatekeeper" for tissue lipid distribution.
Collapse
Affiliation(s)
- Itsaso Garcia-Arcos
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Yaeko Hiyama
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Konstantinos Drosatos
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Kalyani G Bharadwaj
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Yunying Hu
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Ni Huiping Son
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Sheila M O'Byrne
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Chuchun L Chang
- Institute of Human Nutrition, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Richard J Deckelbaum
- Institute of Human Nutrition, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Manabu Takahashi
- Department of Medicine, Jichii University, Tochigi 329-0498, Japan
| | - Marit Westerterp
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032; Department of Medical Biochemistry, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
| | - Joseph C Obunike
- Department of Biological Sciences, New York City College of Technology, City University of New York, Brooklyn, New York 11201
| | - Hongfeng Jiang
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Hiroaki Yagyu
- Department of Medicine, Jichii University, Tochigi 329-0498, Japan
| | - William S Blaner
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032
| | - Ira J Goldberg
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York 10032.
| |
Collapse
|
40
|
The Lipid Transfer Protein StarD7: Structure, Function, and Regulation. Int J Mol Sci 2013; 14:6170-86. [PMID: 23507753 PMCID: PMC3634439 DOI: 10.3390/ijms14036170] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/17/2013] [Accepted: 02/22/2013] [Indexed: 02/06/2023] Open
Abstract
The steroidogenic acute regulatory (StAR) protein-related lipid transfer (START) domain proteins constitute a family of evolutionarily conserved and widely expressed proteins that have been implicated in lipid transport, metabolism, and signaling. The 15 well-characterized mammalian START domain-containing proteins are grouped into six subfamilies. The START domain containing 7 mRNA encodes StarD7, a member of the StarD2/phosphatidylcholine transfer protein (PCTP) subfamily, which was first identified as a gene overexpressed in a choriocarcinoma cell line. Recent studies show that the StarD7 protein facilitates the delivery of phosphatidylcholine to the mitochondria. This review summarizes the latest advances in StarD7 research, focusing on the structural and biochemical features, protein-lipid interactions, and mechanisms that regulate StarD7 expression. The implications of the role of StarD7 in cell proliferation, migration, and differentiation are also discussed.
Collapse
|
41
|
Ardévol A, Motilva MJ, Serra A, Blay M, Pinent M. Procyanidins target mesenteric adipose tissue in Wistar lean rats and subcutaneous adipose tissue in Zucker obese rat. Food Chem 2013; 141:160-6. [PMID: 23768342 DOI: 10.1016/j.foodchem.2013.02.104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Revised: 12/18/2012] [Accepted: 02/25/2013] [Indexed: 12/13/2022]
Abstract
Visceral and subcutaneous adipose depots have different metabolic roles that may be involved in the development of obesity-related pathologies. Procyanidins have beneficial effects on insulin resistance, and they target adipose tissue. We analyse whether procyanidins exert different effects, depending on the adipose tissue depot, and whether these effects show a relation to the amount of phenolic compound in the tissue. We studied the effects of a grape seed procyanidin extract (GSPE) treatment at the transcriptional level on genes expressed differentially between mesenteric and subcutaneous adipose tissue depots and genes previously shown to be targets of procyanidins. Procyanidins target mesenteric adipose tissue in Wistar lean rats but subcutaneous adipose tissue in Zucker obese rats. Non-modified structures also accumulated, preferentially in the same respective tissues that were responsive to GSPE. Thus, procyanidins target and accumulate differently in mesenteric and subcutaneous adipose tissue depots, depending on the metabolic condition of the animal model.
Collapse
Affiliation(s)
- A Ardévol
- Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, C. Marcel·lí Domingo, s/n, 43007 Tarragona, Spain
| | | | | | | | | |
Collapse
|
42
|
Abstract
Excess intra-abdominal adipose tissue accumulation, often termed visceral obesity, is part of a phenotype including dysfunctional subcutaneous adipose tissue expansion and ectopic triglyceride storage closely related to clustering cardiometabolic risk factors. Hypertriglyceridemia; increased free fatty acid availability; adipose tissue release of proinflammatory cytokines; liver insulin resistance and inflammation; increased liver VLDL synthesis and secretion; reduced clearance of triglyceride-rich lipoproteins; presence of small, dense LDL particles; and reduced HDL cholesterol levels are among the many metabolic alterations closely related to this condition. Age, gender, genetics, and ethnicity are broad etiological factors contributing to variation in visceral adipose tissue accumulation. Specific mechanisms responsible for proportionally increased visceral fat storage when facing positive energy balance and weight gain may involve sex hormones, local cortisol production in abdominal adipose tissues, endocannabinoids, growth hormone, and dietary fructose. Physiological characteristics of abdominal adipose tissues such as adipocyte size and number, lipolytic responsiveness, lipid storage capacity, and inflammatory cytokine production are significant correlates and even possible determinants of the increased cardiometabolic risk associated with visceral obesity. Thiazolidinediones, estrogen replacement in postmenopausal women, and testosterone replacement in androgen-deficient men have been shown to favorably modulate body fat distribution and cardiometabolic risk to various degrees. However, some of these therapies must now be considered in the context of their serious side effects. Lifestyle interventions leading to weight loss generally induce preferential mobilization of visceral fat. In clinical practice, measuring waist circumference in addition to the body mass index could be helpful for the identification and management of a subgroup of overweight or obese patients at high cardiometabolic risk.
Collapse
Affiliation(s)
- André Tchernof
- Endocrinology and Genomics Axis, Centre Hospitalier Universitaire de Québec, Québec, Canada
| | | |
Collapse
|
43
|
Miegueu P, St-Pierre DH, Lapointe M, Poursharifi P, Lu H, Gupta A, Cianflone K. Substance P decreases fat storage and increases adipocytokine production in 3T3-L1 adipocytes. Am J Physiol Gastrointest Liver Physiol 2013; 304:G420-7. [PMID: 23257919 DOI: 10.1152/ajpgi.00162.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity, inflammation, and insulin resistance are closely linked. Substance P (SP), via its neurokinin 1 receptor (NK1R), mediates inflammatory and, possibly, neuroendocrine processes. We examined SP effects on lipid storage and cytokine production in 3T3-L1 adipocytes and adipose tissues. 3T3-L1 adipocytes and preadipocytes express NK1R, and 8 days of SP supplementation during differentiation to 3T3-L1 preadipocytes decreased lipid droplet accumulation. SP (10 nM, 24 h) increased lipolysis in primary adipocytes (138 ± 7%, P < 0.05) and reduced fatty acid uptake (-31 ± 7%, P < 0.05) and mRNA expression of the differentiation-related transcription factors peroxisome proliferator-activated receptor-γ type 2 (-64 ± 2%, P < 0.001) and CCAAT enhancer-binding protein (CEBP)-α (-65 ± 2%, P < 0.001) and the lipid storage genes fatty acid-binding protein type 4 (-59 ± 2%, P < 0.001) and diacylglycerol O-acyltransferase-1 (-45 ± 2%, P < 0.01) in 3T3-L1 adipocytes, while CD36, a fatty acid transporter (+82 ± 19%, P < 0.01), was augmented. SP increased secretion of complement C3 (148 ± 15%, P < 0.04), monocyte chemoattractant protein-1 (156 ± 16%, P < 0.03), and keratinocyte-derived chemokine (148 ± 18%, P = 0.045) in 3T3-L1 adipocytes and monocyte chemoattractant protein-1 (496 ± 142%, P < 0.02) and complement C3 (152 ± 25%, P < 0.04) in adipose tissue and primary adipocytes, respectively. These SP effects were accompanied by downregulation of insulin receptor substrate 1 (-82 ± 2%, P < 0.01) and GLUT4 (-76 ± 2%, P < 0.01) mRNA expression, and SP acutely blocked insulin-mediated stimulation of fatty acid uptake and Akt phosphorylation. Although adiponectin secretion was unchanged, mRNA expression was decreased (-86 ± 8%, P < 0.001). In humans, NK1R expression correlates positively with plasma insulin, fatty acid, and complement C3 and negatively with adiponectin, CEBPα, CEBPβ, and peroxisome proliferator-activated receptor-γ mRNA expression in omental, but not subcutaneous, adipose tissue. Our results suggest that, beyond its neuroendocrine and inflammatory effects, SP could also be involved in targeting adipose tissue and influencing insulin resistance.
Collapse
Affiliation(s)
- Pierre Miegueu
- Centre de Recherche Institut Universitaire de Cardiologie and Pneumologie de Quebec and Department of Medicine, Université Laval, Quebec, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
44
|
Miegueu P, St-Pierre DH, Munkonda MN, Lapointe M, Cianflone K. Amylin stimulates fatty acid esterification in 3T3-L1 adipocytes. Mol Cell Endocrinol 2013; 366:99-107. [PMID: 23261986 DOI: 10.1016/j.mce.2012.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 11/20/2012] [Accepted: 12/11/2012] [Indexed: 01/25/2023]
Abstract
Amylin is co-localized and co-secreted with insulin, however its direct effects on adipocytes are unexplored. In 3T3-L1 preadipocytes, amylin increased thymidine incorporation (174%; p<0.05) and Myc mRNA expression (378%; p<0.01). Amylin supplementation during differentiation enhanced triglyceride accumulation (272%; p<0.001). In 3T3-L1 adipocytes, amylin increased fatty acid uptake (238%; p<0.01) and further potentiated the effects of insulin (insulin 158%; p<0.01, amylin+insulin 335%; p<0.001 vs CTL, p<0.001 vs insulin). By contrast, amylin inhibited glycerol release in 3T3-L1 adipocytes (-50%; p<0.05) and primary adipocytes (-34%; p<0.05). Amylin stimulated cytokine secretion (monocyte chemotactic protein-1+166%, keratinocyte-derived chemokine+174%; both p<0.05) and mRNA expression of PPARγ (163%; p<0.01), C/EBPβ (121%, p<0.05), DGAT1 (157%; p<0.01), FABP4 (122%; p<0.01), and CD36 (122%; p<0.05). In human adipose tissue, mRNA expression of amylin receptor genes (CALCR and RAMP3) correlated with numerous lipid and insulin signaling genes, plasma glucose and HOMA. Altogether amylin directly stimulates fat cells, potentiates the effects of insulin and may influence insulin resistance.
Collapse
Affiliation(s)
- Pierre Miegueu
- Centre de Recherche Institut Universitaire de Cardiologie & Pneumologie de Québec, Department of Medicine, Université Laval, Québec, QC, Canada
| | | | | | | | | |
Collapse
|
45
|
Sales V, Patti ME. The Ups and Downs of Insulin Resistance and Type 2 Diabetes: Lessons from Genomic Analyses in Humans. CURRENT CARDIOVASCULAR RISK REPORTS 2012; 7:46-59. [PMID: 23459395 DOI: 10.1007/s12170-012-0283-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We are in the midst of a worldwide epidemic of type 2 diabetes (T2D) and obesity. Understanding the mechanisms underlying these diseases is critical if we are to halt their progression and ultimately prevent their development. The advent and widespread implementation of microarray technology has allowed analysis of small samples of human skeletal muscle, adipose, liver, pancreas and blood. While patterns differ in each tissue, several dominant themes have emerged from these studies, including altered expression of genes indicating increased inflammation and altered lipid and mitochondrial oxidative metabolism and insulin signaling in patients with T2D, and in some cases, in those at risk for disease. Unraveling which changes in gene expression are primary, and which are secondary to an insulin resistant or diabetes metabolic milieu remains a scientific challenge but we are one step closer.
Collapse
Affiliation(s)
- Vicencia Sales
- Research Division, Joslin Diabetes Center, and Department of Medicine, Harvard Medical School ; Department of Biophysics, Federal University of São Paulo, UNIFESP/EPM, São Paulo, SP, Brazil
| | | |
Collapse
|
46
|
van Greevenbroek MMJ, Ghosh S, van der Kallen CJH, Brouwers MCGJ, Schalkwijk CG, Stehouwer CDA. Up-regulation of the complement system in subcutaneous adipocytes from nonobese, hypertriglyceridemic subjects is associated with adipocyte insulin resistance. J Clin Endocrinol Metab 2012; 97:4742-52. [PMID: 23055543 PMCID: PMC3513546 DOI: 10.1210/jc.2012-2539] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Dysfunctional adipose tissue plays an important role in the etiology of the metabolic syndrome, type 2 diabetes, and dyslipidemia. However, the molecular mechanisms underlying adipocyte dysfunction are incompletely understood. AIM The aim of the study was to identify differentially regulated pathways in sc adipocytes of dyslipidemic subjects. METHODS Whole-genome expression profiling was conducted on sc adipocytes from a discovery group of nine marginally overweight subjects with familial combined hyperlipidemia (FCHL) and nine controls of comparable body sizes as well as two independent confirmation groups. In this study, FCHL served as a model of familial insulin resistance and dyslipidemia, in the absence of frank obesity. RESULTS Functional analyses and gene set enrichment analysis using the Kyoto Encyclopedia of Genes and Genomes or a custom pathway database identified the complement system and complement regulators as one of the top up-regulated pathways in FCHL [false discovery rate (FDR) < 1E-30]. Higher adipocyte complement expression in FCHL was confirmed in the appropriate confirmation group. Higher complement gene expression was associated with lower adipocyte insulin receptor substrate-1 expression as marker of adipocyte insulin resistance, independent of age, sex, or disease status, and this association was corroborated in the two confirmation groups. Additionally, complement gene expression was associated with triglycerides in the discovery set and with triglycerides and/or waist circumference in the confirmation groups. Complement pathway up-regulation did not appear to be driven by hypertriglyceridemia because a 40% pharmacological reduction in triglycerides did not affect complement expression. CONCLUSIONS These findings point to an up-regulation of a complement-related transcriptome in sc adipocytes under metabolically stressed conditions, even in the absence of overt obesity. Such up-regulation may subsequently influence downstream processes, including macrophage infiltration into adipose tissue and adipocyte insulin resistance.
Collapse
Affiliation(s)
- M M J van Greevenbroek
- Department of Internal Medicine, Division of General Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
47
|
Kusminski CM, Scherer PE. Mitochondrial dysfunction in white adipose tissue. Trends Endocrinol Metab 2012; 23:435-43. [PMID: 22784416 PMCID: PMC3430798 DOI: 10.1016/j.tem.2012.06.004] [Citation(s) in RCA: 257] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 06/06/2012] [Accepted: 06/11/2012] [Indexed: 12/14/2022]
Abstract
Although mitochondria in brown adipose tissue and their role in non-shivering thermogenesis have been widely studied, we have only a limited understanding of the relevance of mitochondria in white adipose tissue (WAT) for cellular homeostasis of the adipocyte and their impact upon systemic energy homeostasis. A better understanding of the regulatory role that white adipocyte mitochondria play in the regulation of whole-body physiology becomes increasingly important. WAT mitochondrial biogenesis can effectively be induced pharmacologically using a number of agents, including PPARγ agonists. Through their ability to influence key biochemical processes central to the adipocyte, such as fatty acid (FA) esterification and lipogenesis, as well as their impact upon the production and release of key adipokines, mitochondria play a crucial role in determining systemic insulin sensitivity.
Collapse
Affiliation(s)
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, Dallas, Texas 75390-8549
- Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-8549
- corresponding author: , Telephone: (214) 648-8715, Fax: (214) 648-8720
| |
Collapse
|
48
|
Moreno-Navarrete JM, Ortega F, Sánchez-Garrido MÁ, Sabater M, Ricart W, Zorzano A, Tena-Sempere M, Fernández-Real JM. Phosphorylated S6K1 (Thr389) is a molecular adipose tissue marker of altered glucose tolerance. J Nutr Biochem 2012; 24:32-8. [PMID: 22705322 DOI: 10.1016/j.jnutbio.2012.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 12/05/2011] [Accepted: 01/03/2012] [Indexed: 12/25/2022]
Abstract
Molecular tissue markers of altered glucose metabolism will be useful as potential targets for antidiabetic drugs. S6K1 is a downstream signal of insulin action. We aimed to evaluate (pThr389)S6K1 and total S6K1 levels in human and rat fat depots as candidate markers of altered glucose metabolism. (pThr389)S6K1 and total S6K1 levels were measured using enzyme linked immune sorbent assay (ELISA) in 49 adipose tissue samples from subjects with morbid obesity and in 18 peri-renal white adipose tissue samples from rats. The effects of high glucose and rosiglitazone have been explored in human preadipocytes. (pThr389)S6K1/(total)S6K1 in subcutaneous adipose tissue was significantly increased subjects with Type 2 diabetes (0.78 ± 0.26 vs. 0.55 ± 0.14, P=.02) and associated with fasting glucose (r=0.46, P=.04) and glycated hemoglobin (r=0.63, P=.02) in SAT. Similar associations with fasting glucose (r=0.43, P=.03) and IRS1 (r=-0.41, P=.04) gene expression were found in visceral adipose tissue. In addition, rat experiments confirmed the higher (pThr389)S6K1/totalS6K1 levels in adipose tissue in association with obesity-associated metabolic disturbances. (pThr389)S6K1/totalS6K1 was validated using western blot in rat adipose tissue. Both ELISA and western blot data significantly correlated (r=0.85, P=.005). In human preadipocytes, high glucose medium led to increased (pThr389)S6K1/total S6K1 levels in comparison with normal glucose medium, which was significantly decreased under rosiglitazone administration. In conclusion, in human and rat adipose tissue, phosphorylated S6K1 is a marker for increased glucose levels.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBERobn Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III (ISCIII), 17007, Girona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
OBJECTIVE Several gastrointestinal peptides are now recognized to have target functions beyond the intestinal wall, including effects on adipocytes. Secretin (SEC), one of the first identified, has not been evaluated in this context. METHODS Using cultured 3T3-L1 preadipocytes, adipocytes and primary rat adipocytes we evaluated the effect of SEC on cell proliferation, mitochondrial activity, differentiation, triglyceride (TG) synthesis, lipolysis as well expression of the SEC receptor (SCTR) in rodent and human adipose tissues. RESULTS In preadipocytes, SEC significantly increased mitochondrial activity (115%; P<0.01), thymidine incorporation (149.7%; P<0.05) and C/EBPβ expression (123.4%; P<0.05). During standard differentiation, SCTR mRNA increased up to a maximum of ninefold (P<0.001). In human adipose tissue, SCTR correlated with body mass index and plasma insulin, and SCTR mRNA expression was also detected in rat adipose tissues. SEC supplementation during differentiation enhanced TG accumulation (+138%; P<0.01). In mature adipocytes, SEC increased fatty acid (FA) uptake (186%; P<0.01), adiponectin and monocyte chemotactic protein-1 secretion (+142% and +149%, respectively; P<0.05) and mRNA expression of PPARγ (+206%; P<0.01), FABP4 (+164%; P<0.001), DGAT-1 (+144%; P<0.01), adiponectin (+138%; P<0.001) and CD36 (+149%; P<0.05). In primary rat adipocytes, SEC also increased FA uptake (137%; P<0.05). Pretreatment with a SEC antagonist impaired SEC-induced FA uptake and cAMP accumulation. SEC treatment simultaneously stimulated lipolysis measured as glycerol release in 3T3-L1 adipocytes and rat adipose tissue. CONCLUSION The present results suggest that SEC is a potent modulator of adipocyte functions, demonstrating overall a role in enhanced substrate cycling.
Collapse
|
50
|
Kim S, Jwa H, Yanagawa Y, Park T. Extract from Dioscorea batatas ameliorates insulin resistance in mice fed a high-fat diet. J Med Food 2012; 15:527-34. [PMID: 22424459 DOI: 10.1089/jmf.2011.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of this study was to investigate whether Dioscorea batatas (DB) extract attenuates high-fat diet (HFD)-induced insulin resistance in the visceral adipose tissues of mice, and by what mechanism(s). Mice were fed a HFD for 4 weeks to induce the early development of insulin resistance. The DB extract was administered to mice fed a HFD by oral gavage at a dose of 100 mg/kg body weight daily for 7 weeks. Biochemical parameters in blood were measured using enzymatic kits, and the expression levels of glucose transporter 4 (GLUT4), phosphorylated (p-)S6K1, phosphorylated v-akt murine thymoma viral oncogene homolog (p-AKT), and phosphorylated extracellular regulated kinase (p-ERK) in epididymal fat tissue were determined by western blot analyses. The DB extract effectively reversed the HFD-induced elevations in plasma glucose and insulin levels, and the homeostasis model assessment for insulin resistance and oral glucose tolerance test values. The level of p-AKT protein was up-regulated, whereas the levels of p-ERK and p-S6K1 proteins were down-regulated in the adipose tissues of DB mice compared with HFD mice. Furthermore, the DB extract significantly reversed the HFD-induced decrease in the plasma membrane GLUT4 level in the adipose tissue of mice. The DB extract improved glucose metabolism in HFD-fed mice through the up-regulation of plasma membrane GLUT4 content in the visceral adipose tissue. Activation of the insulin signaling cascade leading to GLUT4 translocation was the mechanism underlying the beneficial effects of the DB extract on early-stage obesity-induced insulin resistance.
Collapse
Affiliation(s)
- Soyoung Kim
- Department of Food and Nutrition, Yonsei University, Seoul, Korea
| | | | | | | |
Collapse
|