1
|
Kinin B1 receptor deficiency protects mice fed by cafeteria diet from abnormal glucose homeostasis. PLoS One 2022; 17:e0267845. [PMID: 35617279 PMCID: PMC9135186 DOI: 10.1371/journal.pone.0267845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/14/2022] [Indexed: 11/24/2022] Open
Abstract
The kallikrein–kinin system has been implicated in body weight and glucose homeostasis. Their major effectors act by binding to the kinin B2 and B1 receptors. It was assessed the role of the kinin B1 receptor in weight and glucose homeostasis in B1 receptor knockout mice (B1RKO) subjected to a cafeteria diet (CAF). Wild-type (WT) and B1RKO male mice (C57BL/6 background; 8 weeks old) were fed a standard diet (SD) or CAF for 14 weeks, ad libitum, and four groups were formed: WT-SD; B1RKO-SD; WT-CAF; B1RKO-CAF. Body weight and food intake were assessed weekly. It was performed glucose tolerance (GTT) and insulin tolerance tests (ITT), and HOMA-IR, HOMA-β and HOMA-β* 1/HOMA-IR were calculated. Islets from WT and B1RKO were isolated in order to measure the insulin secretion. Western blot was used to assess the hepatic AKT phosphorylation and qPCR to assess gene expression. CAF induced a higher body mass gain in B1RKO compared to WT mice. CAF diet increased epididymal fat depot mass, hepatic fat infiltration and hepatic AKT phosphorylation in both genotypes. However, B1RKO mice presented lower glycemic response during GTT when fed with CAF, and a lower glucose decrease in the ITT. This higher resistance was overcomed with higher insulin secretion when stimulated by high glucose, resulting in higher glucose uptake in the GTT when submitted to CAF, despite lower insulin sensitivity. Islets from B1RKO delivered 4 times more insulin in 3-month-old mice than islets from WT. The higher insulin disposition index and high insulin delivery of B1RKO can explain the decreased glucose excursion during GTT. In conclusion, CAF increased the β-cell function in B1RKO mice, compensated by the diet-induced insulin resistance and resulting in a healthier glycemic response despite the higher weight gain.
Collapse
|
2
|
Association of Ang-(1–7) and des-Arg9BK as new biomarkers of obesity and cardiometabolic risk factors in adolescents. Hypertens Res 2021; 44:969-977. [DOI: 10.1038/s41440-021-00618-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/26/2020] [Accepted: 12/23/2020] [Indexed: 11/09/2022]
|
3
|
Polidoro RB, Hagan RS, de Santis Santiago R, Schmidt NW. Overview: Systemic Inflammatory Response Derived From Lung Injury Caused by SARS-CoV-2 Infection Explains Severe Outcomes in COVID-19. Front Immunol 2020; 11:1626. [PMID: 32714336 PMCID: PMC7344249 DOI: 10.3389/fimmu.2020.01626] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/17/2020] [Indexed: 01/12/2023] Open
Abstract
Most SARS-CoV2 infections will not develop into severe COVID-19. However, in some patients, lung infection leads to the activation of alveolar macrophages and lung epithelial cells that will release proinflammatory cytokines. IL-6, TNF, and IL-1β increase expression of cell adhesion molecules (CAMs) and VEGF, thereby increasing permeability of the lung endothelium and reducing barrier protection, allowing viral dissemination and infiltration of neutrophils and inflammatory monocytes. In the blood, these cytokines will stimulate the bone marrow to produce and release immature granulocytes, that return to the lung and further increase inflammation, leading to acute respiratory distress syndrome (ARDS). This lung-systemic loop leads to cytokine storm syndrome (CSS). Concurrently, the acute phase response increases the production of platelets, fibrinogen and other pro-thrombotic factors. Systemic decrease in ACE2 function impacts the Renin-Angiotensin-Kallikrein-Kinin systems (RAS-KKS) increasing clotting. The combination of acute lung injury with RAS-KKS unbalance is herein called COVID-19 Associated Lung Injury (CALI). This conservative two-hit model of systemic inflammation due to the lung injury allows new intervention windows and is more consistent with the current knowledge.
Collapse
Affiliation(s)
- Rafael B. Polidoro
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Robert S. Hagan
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | | | - Nathan W. Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
4
|
Ji B, Wang Q, Xue Q, Li W, Li X, Wu Y. The Dual Role of Kinin/Kinin Receptors System in Alzheimer's Disease. Front Mol Neurosci 2019; 12:234. [PMID: 31632239 PMCID: PMC6779775 DOI: 10.3389/fnmol.2019.00234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/13/2019] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease characterized by progressive spatial disorientation, learning and memory deficits, responsible for 60%–80% of all dementias. However, the pathological mechanism of AD remains unknown. Numerous studies revealed that kinin/kinin receptors system (KKS) may be involved in the pathophysiology of AD. In this review article, we summarized the roles of KKS in neuroinflammation, cerebrovascular impairment, tau phosphorylation, and amyloid β (Aβ) generation in AD. Moreover, we provide new insights into the mechanistic link between KKS and AD, and highlight the KKS as a potential therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Bingyuan Ji
- Neurobiology Institute, School of Mental Health, Jining Medical University, Jining, China
| | - Qinqin Wang
- Neurobiology Institute, School of Mental Health, Jining Medical University, Jining, China
| | - Qingjie Xue
- Department of Pathogenic Biology, Jining Medical University, Jining, China
| | - Wenfu Li
- Neurobiology Institute, School of Mental Health, Jining Medical University, Jining, China
| | - Xuezhi Li
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Yili Wu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| |
Collapse
|
5
|
Sales VM, Gonçalves-Zillo T, Castoldi A, Burgos M, Branquinho J, Batista C, Oliveira V, Silva E, Castro CHM, Câmara N, Mori MA, Pesquero JB. Kinin B 1 Receptor Acts in Adipose Tissue to Control Fat Distribution in a Cell-Nonautonomous Manner. Diabetes 2019; 68:1614-1623. [PMID: 31167880 DOI: 10.2337/db18-1150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 05/28/2019] [Indexed: 11/13/2022]
Abstract
The kinin B1 receptor (B1R) plays a role in inflammatory and metabolic processes. B1R deletion (B1 -/-) protects mice from diet-induced obesity and improves insulin and leptin sensitivity. In contrast, genetic reconstitution of B1R exclusively in adipose tissue reverses the lean phenotype of B1 -/- mice. To study the cell-nonautonomous nature of these effects, we transplanted epididymal white adipose tissue (eWAT) from wild-type donors (B1 +/+) into B1 -/- mice (B1 +/+→B1 -/-) and compared them with autologous controls (B1 +/+→B1 +/+ or B1 -/-→B1 -/-). We then fed these mice a high-fat diet for 16 weeks and investigated their metabolic phenotypes. B1 +/+→B1 -/- mice became obese but not glucose intolerant or insulin resistant, unlike B1 -/-→B1 -/- mice. Moreover, the endogenous adipose tissue of B1 +/+→B1 -/- mice exhibited higher expression of adipocyte markers (e.g., Fabp4 and Adipoq) and changes in the immune cell pool. These mice also developed fatty liver. Wild-type eWAT transplanted into B1 -/- mice normalized circulating insulin, leptin, and epidermal growth factor levels. In conclusion, we demonstrated that B1R in adipose tissue controls the response to diet-induced obesity by promoting adipose tissue expansion and hepatic lipid accumulation in cell-nonautonomous manners.
Collapse
Affiliation(s)
- Vicencia M Sales
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Thais Gonçalves-Zillo
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Angela Castoldi
- Department of Immunology, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Marina Burgos
- Department of Immunology, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Jessica Branquinho
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Carolina Batista
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Valeria Oliveira
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Elton Silva
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Charlles H M Castro
- Department of Medicine, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Niels Câmara
- Department of Immunology, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Marcelo A Mori
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Department of Biochemistry and Tissue Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - João Bosco Pesquero
- Department of Biophysics, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
6
|
Ding R, Yang M, Quan J, Li S, Zhuang Z, Zhou S, Zheng E, Hong L, Li Z, Cai G, Huang W, Wu Z, Yang J. Single-Locus and Multi-Locus Genome-Wide Association Studies for Intramuscular Fat in Duroc Pigs. Front Genet 2019; 10:619. [PMID: 31316554 PMCID: PMC6609572 DOI: 10.3389/fgene.2019.00619] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 06/13/2019] [Indexed: 12/26/2022] Open
Abstract
Intramuscular fat (IMF) is an important quantitative trait of meat, which affects the associated sensory properties and nutritional value of pork. To gain a better understanding of the genetic determinants of IMF, we used a composite strategy, including single-locus and multi-locus association analyses to perform genome-wide association studies (GWAS) for IMF in 1,490 Duroc boars. We estimated the genomic heritability of IMF to be 0.23 ± 0.04. A total of 30 single nucleotide polymorphisms (SNPs) were found to be significantly associated with IMF. The single-locus mixed linear model (MLM) and multiple-locus methods multi-locus random-SNP-effect mixed linear model (mrMLM), fast multi-locus random-SNP-effect efficient mixed model association (FASTmrEMMA), and integrative sure independence screening expectation maximization Bayesian least absolute shrinkage and selection operator model (ISIS EM-BLASSO) analyses identified 5, 9, 8, and 21 significant SNPs, respectively. Interestingly, a novel quantitative trait locus (QTL) on SSC 7 was found to affect IMF. In addition, 10 candidate genes (BDKRB2, GTF2IRD1, UTRN, TMEM138, DPYD, CASQ2, ZNF518B, S1PR1, GPC6, and GLI1) were found to be associated with IMF based on their potential functional roles in IMF. GO analysis showed that most of the genes were involved in muscle and organ development. A significantly enriched KEGG pathway, the sphingolipid signaling pathway, was reported to be associated with fat deposition and obesity. Identification of novel variants and functional genes will advance our understanding of the genetic mechanisms of IMF and provide specific opportunities for marker-assisted or genomic selection in pigs. In general, such a composite single-locus and multi-locus strategy for GWAS may be useful for understanding the genetic architecture of economic traits in livestock.
Collapse
Affiliation(s)
- Rongrong Ding
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Ming Yang
- National Engineering Research Center for Breeding Swine Industry, Guangdong Wens Foodstuffs Group, Co., Ltd., Guangdong, China
| | - Jianping Quan
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Shaoyun Li
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Zhanwei Zhuang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Shenping Zhou
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Enqin Zheng
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Linjun Hong
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Zicong Li
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| | - Gengyuan Cai
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China.,National Engineering Research Center for Breeding Swine Industry, Guangdong Wens Foodstuffs Group, Co., Ltd., Guangdong, China
| | - Wen Huang
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
| | - Zhenfang Wu
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China.,National Engineering Research Center for Breeding Swine Industry, Guangdong Wens Foodstuffs Group, Co., Ltd., Guangdong, China
| | - Jie Yang
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangdong, China
| |
Collapse
|
7
|
Meng Q, Ge S, Yan W, Li R, Dou J, Wang H, Wang B, Ma Q, Zhou Y, Song M, Yu X, Wang H, Yang X, Liu F, Alzain MA, Yan Y, Zhang L, Wu L, Zhao F, He Y, Guo X, Chen F, Xu W, Garcia M, Menon D, Wang Y, Mu Y, Wang W. Screening for potential serum-based proteomic biomarkers for human type 2 diabetes mellitus using MALDI-TOF MS. Proteomics Clin Appl 2016; 11. [PMID: 27863080 DOI: 10.1002/prca.201600079] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 10/04/2016] [Accepted: 11/04/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Qiutao Meng
- Department of Endocrinology; Chinese PLA General Hospital; Beijing China
| | - Siqi Ge
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
- School of Medical and Health Sciences; Edith Cowan University; Perth Australia
| | - Wenhua Yan
- Department of Endocrinology; Chinese PLA General Hospital; Beijing China
| | - Ruisheng Li
- Research and Technology Service Center; Chinese PLA 302 Hospital Beijing China
| | - Jingtao Dou
- Department of Endocrinology; Chinese PLA General Hospital; Beijing China
| | - Haibing Wang
- Department of Endocrinology; Chinese PLA General Hospital; Beijing China
| | - Baoan Wang
- Department of Endocrinology; Chinese PLA General Hospital; Beijing China
| | - Qingwei Ma
- Bioyong (Beijing) Technology Co., Ltd.; Beijing China
| | - Yong Zhou
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Manshu Song
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Xinwei Yu
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
- School of Medical and Health Sciences; Edith Cowan University; Perth Australia
| | - Hao Wang
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Xinghua Yang
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Fen Liu
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Mohamed Ali Alzain
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Yuxiang Yan
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Ling Zhang
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Lijuan Wu
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Feifei Zhao
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Yan He
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Xiuhua Guo
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Feng Chen
- Central of Laboratory; Peking University School and Hospital of Stomatology; Beijing China
| | - Weizhuo Xu
- School of Life Science and Biopharmaceuticals; Shenyang Pharmaceutical University; Shenyang China
| | - Monique Garcia
- School of Medical and Health Sciences; Edith Cowan University; Perth Australia
| | - Desmond Menon
- School of Medical and Health Sciences; Edith Cowan University; Perth Australia
| | - Youxin Wang
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
| | - Yiming Mu
- Department of Endocrinology; Chinese PLA General Hospital; Beijing China
| | - Wei Wang
- Beijing Key Laboratory of Clinical Epidemiology; School of Public Health; Capital Medical University; Beijing China
- School of Medical and Health Sciences; Edith Cowan University; Perth Australia
| |
Collapse
|
8
|
Breit A, Besik V, Solinski HJ, Muehlich S, Glas E, Yarwood SJ, Gudermann T. Serine-727 phosphorylation activates hypothalamic STAT-3 independently from tyrosine-705 phosphorylation. Mol Endocrinol 2015; 29:445-59. [PMID: 25584415 DOI: 10.1210/me.2014-1300] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Transcriptional activity of signal transducer and activator of transcription-3 (STAT-3) is a key element in the central regulation of appetite and energy homeostasis. Activation of hypothalamic STAT-3 has been attributed to cytokine-promoted phosphorylation at tyrosine-705 (Tyr-705). In nonhypothalamic cells, STAT-3 is also phosphorylated at serine-727 (Ser-727), but the functional significance of Ser-727 in the regulation of hypothalamic STAT-3 is not known. We used 2 hypothalamic cell lines and analyzed the effects of various hormones on STAT-3-dependent reporter gene activity and observed that IFN-γ, epidermal growth factor (EGF), and bradykinin (BK) induce similar STAT-3 reporter activation. EGF and BK solely increased Ser-727 and IFN-γ increased Tyr-705 phosphorylation of STAT-3. Specific inhibition of ERK-1/2 activity blocked EGF- and BK-induced STAT-3 activation and Ser-727 phosphorylation. BK-induced ERK-1/2 activation occurred via EGF receptor transactivation. Consequently, the BK-mediated effects on STAT-3 were blocked by a specific EGF receptor antagonist. Next, we analyzed the effects of IFN-γ and EGF on the expression of the STAT-3-dependent genes thyroliberin-releasing hormone and suppressors of cytokine signaling-3. EGF but not IFN-γ enhanced thyroliberin-releasing hormone expression via STAT-3. With regard to suppressors of cytokine signaling-3, we observed prolonged expression induced by IFN-γ and a transient effect of EGF that required coactivation of the activator protein-1. Thus, EGF-promoted Ser-727 phosphorylation by ERK-1/2 is not only sufficient to fully activate hypothalamic STAT-3, but, in terms of targeted genes and required cofactors, entails distinct modes of STAT-3 actions compared with IFN-γ-induced Tyr-705 phosphorylation.
Collapse
Affiliation(s)
- Andreas Breit
- Walther-Straub-Institut für Pharmakologie und Toxikologie (A.B., V.B., H.J.S., S.M., E.G., T.G.), Ludwig-Maximilians-Universität München, München, Germany 80336; and The Institute of Molecular, Cell and Systems Biology (S.J.Y.), College of Medical, Veterinary and Life Science, University of Glasgow, Glasgow GC12 8QQ, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
9
|
Midaoui AE, Talbot S, Lahjouji K, Dias JP, Fantus IG, Couture R. Effects of Alpha-Lipoic Acid on Oxidative Stress and Kinin Receptor Expression in Obese Zucker Diabetic Fatty Rats. ACTA ACUST UNITED AC 2015; 6:1-7. [PMID: 26413386 PMCID: PMC4580416 DOI: 10.4172/2155-6156.1000556] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objective To investigate the impact of alpha-lipoic acid on superoxide anion production and NADPH oxidase activity as well as on the expression of kinin B1 and B2 receptors in key organs of obese Zucker Diabetic Fatty rats. Methods Superoxide anion production was measured by lucigenin chemiluminescence. Kinin B1 and B2 receptors expression was measured at protein and mRNA levels by western blot and qRT-PCR in key organs of Zucker Diabetic Fatty and Zucker lean control rats treated for a period of 6 weeks with a standard diet or a diet containing the antioxidant α-lipoic acid (1 g/kg). Results Superoxide anion production and NADPH oxidase activity were significantly enhanced in aorta and adipose tissue of Zucker Diabetic Fatty rats. Kinin B1 and B2 receptors expression levels were also significantly increased in the liver and the gastrocnemius muscle of Zucker Diabetic Fatty rats. Expression of both receptors was not altered in the pancreas of Zucker Diabetic Fatty rats and was undetectable in white retroperitoneal adipose tissue. Alpha-lipoic acid prevented the rise in NADPH oxidase activity in aorta and epididymal adipose tissue of Zucker Diabetic Fatty rats and the upregulation of kinin B1 receptor in liver and gastrocnemius muscle and that of kinin B2 receptor in the liver. Alpha-lipoic acid treatment was found to prevent the final body weight increase without affecting significantly hyperglycemia, hyperinsulinemia and insulin resistance index in Zucker Diabetic Fatty rats. Conclusion Findings support the hypothesis that oxidative stress is implicated in the induction of kinin B1 receptor in Zucker Diabetic Fatty rats. The ability of α-lipoic acid to blunt the body weight gain appears to be mediated in part by preventing NADPH oxidase activity rise in adipose tissue and reversing the hepatic upregulation of kinin B1 receptor in Zucker Diabetic Fatty rats.
Collapse
Affiliation(s)
- Adil El Midaoui
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, PO Box 6128, Station City-Center, Montréal, Qc, H3C 3J7 Canada
| | - Sébastien Talbot
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, PO Box 6128, Station City-Center, Montréal, Qc, H3C 3J7 Canada
| | - Karim Lahjouji
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, PO Box 6128, Station City-Center, Montréal, Qc, H3C 3J7 Canada
| | - Jenny Pena Dias
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, PO Box 6128, Station City-Center, Montréal, Qc, H3C 3J7 Canada
| | - I George Fantus
- Department of Medicine, Mount Sinai Hospital and University Health Network, Banting and Best Diabetes Center, University of Toronto, Toronto, On, M5G 2C4 Canada
| | - Réjean Couture
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, PO Box 6128, Station City-Center, Montréal, Qc, H3C 3J7 Canada
| |
Collapse
|
10
|
Marcadenti A. <i>ADRB2</i>, <i>ADRB3</i>, <i>BDKRB2</i> and <i>MTNR1B</i> Genes Related to Body fat Modulation and Its Interaction with Physical Activity and Blood Pressure. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/ojemd.2015.57012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
11
|
LC-MS/MS analysis of visceral and subcutaneous adipose tissue proteomes in young goats with focus on innate immunity and inflammation related proteins. J Proteomics 2014; 108:295-305. [PMID: 24911890 DOI: 10.1016/j.jprot.2014.05.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 05/27/2014] [Accepted: 05/28/2014] [Indexed: 11/22/2022]
Abstract
UNLABELLED The endocrine role of adipose tissue and its involvement in several physiological and pathological processes are well recognized. Studies on human, mouse and rat adipose tissues have made clear that subcutaneous and visceral deposits play different roles, which is also reflected by different protein and gene expression patterns. In ruminants, fat tissues play important biological roles not only for animal health, but also for quality and gain in meat and milk production. Yet very few studies have explored the ruminant adipose tissue proteomes. The aim of our study was to compare subcutaneous and visceral adipose tissues of goat, focusing on proteins involved in immune and inflammatory response. A 2-D LC-MS/MS approach followed by cluster analysis shows a clear distinction between subcutaneous and visceral fat tissue proteomes, and qualitative RT-PCR based analysis of 30 potential adipokines further confirmed the individual expression patterns of 26 of these, including 7 whose mRNA expression was observed for the first time in adipose tissues. This study provides a first description of adipose tissue proteomes in goat, and presents observations on novel proteins related to metabolic and inflammatory pathways. The mass spectrometry data have been deposited to the ProteomeXchange Consortium with the data set identifier PXD000564. BIOLOGICAL SIGNIFICANCE The proteomic analysis of different subcutaneous and visceral adipose tissue deposits showed tissue specific differences in protein expressions of well known as well as novel adipokines. This highlights the importance of sampling site when studying adipose tissue's metabolic roles. The protein expression characteristics of adipose tissues was evaluated by quantitative RT-PCR, and confirmed that adipose tissues play a central role in controlling inflammation, detoxification and coagulation pathways, as well as regulation of body fat mobilization in dairy animals. These findings are of particular interest in farm animals where health and production traits are important for animal welfare and for economic gains.
Collapse
|
12
|
Rodrigues ES, Martin RP, Silva RF, Nakaie CR, Oliveira L, Shimuta SI. Binding affinities and activation of Asp712Ala and Cys100Ser mutated kinin B1 receptor forms suggest a bimodal scheme for the molecule of bound-DABK. REGULATORY PEPTIDES 2013; 181:37-44. [PMID: 23318500 DOI: 10.1016/j.regpep.2012.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 11/30/2012] [Accepted: 12/17/2012] [Indexed: 06/01/2023]
Abstract
Mutant forms of kinin B(1) receptor (B(1)R) and analogs of the full agonist des-Arg(9)-bradykinin (DABK) were investigated aiming to verify the importance of selected receptor residues and of each agonist-peptide residue in the specific binding and activation. Linked by a specific disulfide bond (Cys(100)-Cys(650)), the N-terminal (N(t)) and the EC3 loop C-terminal (C(t)) segments of angiotensin II (AngII) receptor 1 (AT(1)R) have been identified to form an extracellular site for binding the agonist N(t) segment (Asp(1) and Arg(2) residues). Asp(712) residue at the receptor EC3 loop binds the peptide Arg(2) residue. By homology, a similar site might be considered for DABK binding to B(1)R since this receptor contains the same structural elements for composing the site in AT(1)R, namely the disulfide bond and the EC3 loop Asp(712) residue. DABK, Ala(n)-DABK analogs (n=Ala(1)-, Ala(2)-, Ala(3)-, Ala(4)-, Ala(5)-, Ala(6)-, Ala(7)-, Ala(8)-DABK), and other analogs were selected to binding wild-type, Asp712Ala and Cys100Ser mutated B(1)R receptors. The results obtained suggested that the same bimodal scheme adopted for AngII-AT(1)R system may be applied to DABK binding to B(1)R. The most crucial similarity in the two cases is that the N(t) segments of peptides equally bind to the homologous Asp(712) residue of both AT(1)R and B(1)R extracellular sites. Confirming this preliminary supposition, mutation of residues located at the B(1)R extracellular site as EC3 loop Asp(712) and Cys(100) caused the same modifications in biological assays observed in AT(1)R submitted to homologous mutations, such as significant weakening of agonist binding and reduction of post-receptor-activation processes. These findings provided enough support for defining a site that determines the specific binding of DABK to B(1)R receptors.
Collapse
Affiliation(s)
- E S Rodrigues
- Biophysics Department, Universidade Federal de São Paulo, Rua Botucatu, 862 7th floor, São Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
13
|
Lack of kinin B1 receptor potentiates leptin action in the liver. J Mol Med (Berl) 2013; 91:851-60. [DOI: 10.1007/s00109-013-1004-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 01/08/2013] [Accepted: 01/21/2013] [Indexed: 12/18/2022]
|
14
|
Simon J, Milenkovic D, Godet E, Cabau C, Collin A, Métayer-Coustard S, Rideau N, Tesseraud S, Derouet M, Crochet S, Cailleau-Audouin E, Hennequet-Antier C, Gespach C, Porter TE, Duclos MJ, Dupont J, Cogburn LA. Insulin immuno-neutralization in fed chickens: effects on liver and muscle transcriptome. Physiol Genomics 2012; 44:283-92. [PMID: 22214599 DOI: 10.1152/physiolgenomics.00057.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chickens mimic an insulin-resistance state by exhibiting several peculiarities with regard to plasma glucose level and its control by insulin. To gain insight into the role of insulin in the control of chicken transcriptome, liver and leg muscle transcriptomes were compared in fed controls and "diabetic" chickens, at 5 h after insulin immuno-neutralization, using 20.7K-chicken oligo-microarrays. At a level of false discovery rate <0.01, 1,573 and 1,225 signals were significantly modified by insulin privation in liver and muscle, respectively. Microarray data agreed reasonably well with qRT-PCR and some protein level measurements. Differentially expressed mRNAs with human ID were classified using Biorag analysis and Ingenuity Pathway Analysis. Multiple metabolic pathways, structural proteins, transporters and proteins of intracellular trafficking, major signaling pathways, and elements of the transcriptional control machinery were largely represented in both tissues. At least 42 mRNAs have already been associated with diabetes, insulin resistance, obesity, energy expenditure, or identified as sensors of metabolism in mice or humans. The contribution of the pathways presently identified to chicken physiology (particularly those not yet related to insulin) needs to be evaluated in future studies. Other challenges include the characterization of "unknown" mRNAs and the identification of the steps or networks, which disturbed tissue transcriptome so extensively, quickly after the turning off of the insulin signal. In conclusion, pleiotropic effects of insulin in chickens are further evidenced; major pathways controlled by insulin in mammals have been conserved despite the presence of unique features of insulin signaling in chicken muscle.
Collapse
Affiliation(s)
- Jean Simon
- Station de Recherches Avicoles, INRA, 37380 Nouzilly, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Okada S, List EO, Sankaran S, Kopchick JJ. Plasma Protein Biomarkers Correlated with the Development of Diet-Induced Type 2 Diabetes in Mice. Clin Proteomics 2010; 6:6-17. [PMID: 20625478 DOI: 10.1007/s12014-009-9040-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION: Early detection, assessment of disease progression, and application of an appropriate therapeutic intervention are all important for the care of patients with type 2 diabetes. Currently, however, there is no simple test for early detection of type 2 diabetes. Established diagnostic tests for the disease including oral glucose tolerance, fasting blood glucose, and hemoglobin A1c are relatively late markers where the disease has already progressed. Since blood is in direct contact with many tissues, we hypothesized that pathological tissue changes are likely to be reflected in proteomic profiles of plasma. METHODS: Mice were reared either on regular chow or a high-fat diet at weaning and several physiological responses (i.e., weight, fasting plasma glucose and insulin, and glucose tolerance) were monitored at regular time intervals. Plasma was collected at regular intervals for proteomic analysis by two-dimensional gel electrophoresis and subsequent mass spectrometry. RESULTS: Onset of hyperinsulinemia with corresponding glucose intolerance was observed in 2 weeks and fasting blood glucose levels rose significantly after 4 weeks on the high-fat diet. Many proteins were found to exist in multiple forms (isoforms). Levels of some isoforms including plasma retinol binding protein, transthyretin, Apolipoprotein A1, and kininogen showed significant changes as early as 4 weeks which coincided with the very early development of glucose intolerance. CONCLUSIONS: These results show that a proteomic approach to study the development of type 2 diabetes may uncover unknown early post-translationally modified diagnostic and/or therapeutic protein targets.
Collapse
Affiliation(s)
- Shigeru Okada
- Edison Biotechnology Institute, Konneker Research Laboratories, Ohio University, The Ridges, Bldg. 25, Athens, OH 45701-2979, USA, Department of Pediatrics, College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | | | | | | |
Collapse
|
16
|
Merino VF, Todiras M, Mori MA, Sales VMT, Fonseca RG, Saul V, Tenner K, Bader M, Pesquero JB. Predisposition to atherosclerosis and aortic aneurysms in mice deficient in kinin B1 receptor and apolipoprotein E. J Mol Med (Berl) 2009; 87:953-63. [PMID: 19618151 DOI: 10.1007/s00109-009-0501-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 05/20/2009] [Accepted: 06/29/2009] [Indexed: 01/11/2023]
Abstract
Kinin B1 receptor is involved in chronic inflammation and expressed in human atherosclerotic lesions. However, its significance for lesion development is unknown. Therefore, we investigated the effect of kinin B1 receptor deletion on the development of atherosclerosis and aortic aneurysms in apolipoprotein E-deficient (ApoE(-/-)) mice. Mice deficient both in ApoE and in kinin B1 receptor (ApoE(-/-)-B(1)(-/-)) were generated and analyzed for their susceptibility to atherosclerosis and aneurysm development under cholesterol rich-diet (western diet) and angiotensin II infusion. Kinin B1 receptor messenger RNA (mRNA) expression was significantly increased in ApoE(-/-) mice after Western-type diet. Although no difference in serum cholesterol was found between ApoE(-/-)-B(1)(-/-) and ApoE(-/-) mice under Western-type diet, aortic lesion incidence was significantly higher in ApoE(-/-)-B(1)(-/-) after this treatment. In accordance, we observed increased endothelial dysfunction in these mice. The mRNA expression of cyclic guanosine monophosphate-dependent protein kinase I, CD-11, F4/80, macrophage colony-stimulating factor, and tumor necrosis factor-alpha were increased in the aorta of double-deficient mice following Western-type diet, whereas the levels of peroxisome proliferator-activated receptor gamma protein and the activity of matrix metalloproteinase-9 activity were decreased. In addition to the increased atherosclerotic lesions, the lack of kinin B(1) receptor also increased the incidence of abdominal aortic aneurysms after angiotensin II infusion. In conclusion, our results show that kinin B(1) receptor deficiency aggravates atherosclerosis and aortic aneurysms under cholesterolemic conditions, supporting an antiatherogenic role for the kinin B(1) receptor.
Collapse
Affiliation(s)
- Vanessa F Merino
- Department of Biophysics, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Mori MA, Araújo RC, Reis FCG, Sgai DG, Fonseca RG, Barros CC, Merino VF, Passadore M, Barbosa AM, Ferrari B, Carayon P, Castro CHM, Shimuta SI, Luz J, Bascands JL, Schanstra JP, Even PC, Oliveira SM, Bader M, Pesquero JB. Kinin B1 receptor deficiency leads to leptin hypersensitivity and resistance to obesity. Diabetes 2008; 57:1491-500. [PMID: 18332096 DOI: 10.2337/db07-1508] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Kinins mediate pathophysiological processes related to hypertension, pain, and inflammation through the activation of two G-protein-coupled receptors, named B(1) and B(2). Although these peptides have been related to glucose homeostasis, their effects on energy balance are still unknown. RESEARCH DESIGN AND METHODS Using genetic and pharmacological strategies to abrogate the kinin B(1) receptor in different animal models of obesity, here we present evidence of a novel role for kinins in the regulation of satiety and adiposity. RESULTS Kinin B(1) receptor deficiency in mice (B(1)(-/-)) resulted in less fat content, hypoleptinemia, increased leptin sensitivity, and robust protection against high-fat diet-induced weight gain. Under high-fat diet, B(1)(-/-) also exhibited reduced food intake, improved lipid oxidation, and increased energy expenditure. Surprisingly, B(1) receptor deficiency was not able to decrease food intake and adiposity in obese mice lacking leptin (ob/ob-B(1)(-/-)). However, ob/ob-B(1)(-/-) mice were more responsive to the effects of exogenous leptin on body weight and food intake, suggesting that B(1) receptors may be dependent on leptin to display their metabolic roles. Finally, inhibition of weight gain and food intake by B(1) receptor ablation was pharmacologically confirmed by long-term administration of the kinin B(1) receptor antagonist SSR240612 to mice under high-fat diet. CONCLUSIONS Our data suggest that kinin B(1) receptors participate in the regulation of the energy balance via a mechanism that could involve the modulation of leptin sensitivity.
Collapse
Affiliation(s)
- Marcelo A Mori
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Buléon M, Allard J, Jaafar A, Praddaude F, Dickson Z, Ranera MT, Pecher C, Girolami JP, Tack I. Pharmacological blockade of B2-kinin receptor reduces renal protective effect of angiotensin-converting enzyme inhibition in db/db mice model. Am J Physiol Renal Physiol 2008; 294:F1249-56. [PMID: 18367657 DOI: 10.1152/ajprenal.00501.2007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetic nephropathy (DN) can be delayed by the use of angiotensin-converting enzyme inhibitors (ACEi). The mechanisms of ACEi renal protection are not univocal. To investigate the impact of bradykinin B(2) receptor (B2R) activation during ACE inhibition, type II diabetic mice (C57BLKS db/db) received for 20 wk: 1) ACEi (ramipril) alone, 2) ACEi + HOE-140 (a specific B2R antagonist), 3) HOE-140 alone, or 4) no treatment. The development of DN, defined by an increase in albuminuria and glomerulosclerosis, was largely prevented by ACEi treatment (albuminuria: 980 +/- 130 vs. 2,160 +/- 330 mg/g creatinine; mesangial area: 22.5 +/- 0.5 vs. 27.6 +/- 0.3%). The protective effect of ramipril was markedly attenuated by B2R blockade (albuminuria: 2,790 +/- 680 mg/g creatinine; mesangial area: 30.4 +/- 1.1%), whereas HOE-140 alone significantly increased albuminuria. Despite such benefits, glomerular filtration rate remained unchanged, probably because of the combination of the hypotensive effect of diabetes in this model and the renal hemodynamic action of ramipril. Finally, the renal protective effect of ACEi was associated with a marked decrease in glomerular overexpression of insulin-like growth factor-1 (IGF-1) and transforming growth factor-beta pathways, but also in advanced glycation end product receptors and lipid peroxidation assessed by 4-hydroxy-2-nonenal (4-HNE) adducts. Concomitant blockade of B2R partly restored glomerular overexpression of IGF-1 receptor beta and 4-HNE complexes. These results support the critical role of B2R activation in the mediation of ACEi renal protection against DN and provide the rationale to examine the benefit of B2R activation by itself as a new therapeutic approach for DN.
Collapse
Affiliation(s)
- Marie Buléon
- Laboratoire de Physiologie, Faculté de Médecine Rangueil, Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kinin B1 receptor stimulation modulates leptin homeostasis. Evidence for an insulin-dependent mechanism. Int Immunopharmacol 2008; 8:242-6. [DOI: 10.1016/j.intimp.2007.07.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2007] [Accepted: 07/27/2007] [Indexed: 11/22/2022]
|