1
|
Monteiro BL, Santos RAS, Mario EG, Araujo TS, Savergnini SSQ, Santiago AF, Muzzi RAL, Castro IC, Teixeira LG, Botion LM, Marinho BM, Santos SHS, Porto LCJ. Genetic deletion of Mas receptor in FVB/N mice impairs cardiac use of glucose and lipids. Peptides 2022; 151:170764. [PMID: 35151766 DOI: 10.1016/j.peptides.2022.170764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/18/2022] [Accepted: 02/08/2022] [Indexed: 11/20/2022]
Abstract
Angiotensin-(1-7) is a biologically active product of the renin-angiotensin system cascade and exerts inhibitory effects on inflammation, vascular and cellular growth mechanisms signaling through the G protein-coupled Mas receptor. The major purpose of the present study was to investigate the use of glucose and fatty acids by cardiac tissue in Mas knockout mice models. Serum levels of glucose, lipids, and insulin were measured in Mas-deficient and wild-type FVB/N mice. To investigate the cardiac use of lipids, the lipoprotein lipase, the gene expression of peroxisome proliferator-activated receptor alpha; carnitine palmitoyltransferase I and acyl-CoA oxidase were evaluated. To investigate the cardiac use of glucose, the insulin signaling through Akt/GLUT4 pathway, glucose-6-phosphate (G-6-P) and fructose-6-phosphate (F-6-P) glycolytic intermediates, in addition to ATP, lactate and the glycogen content were measured. Despite normal body weight, cholesterol and insulin, Mas-Knockout mice presented hyperglycemia and hypertriglyceridemia, impaired insulin signaling, through reduced phosphorylation of AKT and decreased translocation of GLUT4 in response to insulin, with subsequent decrease of the cardiac G-6-P and F-6-P. Lactate production and glycogen content were not altered in Mas-KO hearts. Mas-KO presented reduced cardiac lipoprotein lipase activity and decreased translocation of CD36 in response to insulin. The expression of peroxisome proliferator-activated receptor alpha and carnitine palmitoyltransferase I genes were lower in Mas-KO animals compared to wild-type animals. The ATP content of Mas-KO hearts was smaller than in wild-type. The present results suggest that genetic deletion of Mas produced a devastating effect on cardiac use of glucose and lipids, leading to lower energy efficiency in the heart.
Collapse
Affiliation(s)
- Brenda L Monteiro
- Federal University of Lavras, Department of Nutrition, Av. Norte UFLA - Aquenta Sol, Lavras, MG, Brazil.
| | - Robson A S Santos
- Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, Brazil.
| | - Erica G Mario
- Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, Brazil.
| | - Thiago S Araujo
- Federal University of Lavras, Department of Nutrition, Av. Norte UFLA - Aquenta Sol, Lavras, MG, Brazil.
| | - Silvia S Q Savergnini
- Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, Brazil.
| | - Andrezza F Santiago
- Federal University of Lavras, Department of Nutrition, Av. Norte UFLA - Aquenta Sol, Lavras, MG, Brazil.
| | - Ruthnea A L Muzzi
- Institute of Agricultural Sciences (ICA), Food Engineering, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| | - Isabela C Castro
- Federal University of Lavras, Department of Nutrition, Av. Norte UFLA - Aquenta Sol, Lavras, MG, Brazil.
| | - Lilian G Teixeira
- Federal University of Lavras, Department of Nutrition, Av. Norte UFLA - Aquenta Sol, Lavras, MG, Brazil.
| | - Leida M Botion
- Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, Brazil.
| | - Barbhara M Marinho
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil.
| | - Sergio H S Santos
- Institute of Agricultural Sciences (ICA), Food Engineering, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil; Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Minas Gerais, Brazil.
| | - Laura C J Porto
- Federal University of Lavras, Department of Nutrition, Av. Norte UFLA - Aquenta Sol, Lavras, MG, Brazil; Institute of Biological Sciences, Department of Physiology and Biophysics, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, Brazil.
| |
Collapse
|
2
|
Tuleta I, Frangogiannis NG. Diabetic fibrosis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166044. [PMID: 33378699 PMCID: PMC7867637 DOI: 10.1016/j.bbadis.2020.166044] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/25/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022]
Abstract
Diabetes-associated morbidity and mortality is predominantly due to complications of the disease that may cause debilitating conditions, such as heart and renal failure, hepatic insufficiency, retinopathy or peripheral neuropathy. Fibrosis, the excessive and inappropriate deposition of extracellular matrix in various tissues, is commonly found in patients with advanced type 1 or type 2 diabetes, and may contribute to organ dysfunction. Hyperglycemia, lipotoxic injury and insulin resistance activate a fibrotic response, not only through direct stimulation of matrix synthesis by fibroblasts, but also by promoting a fibrogenic phenotype in immune and vascular cells, and possibly also by triggering epithelial and endothelial cell conversion to a fibroblast-like phenotype. High glucose stimulates several fibrogenic pathways, triggering reactive oxygen species generation, stimulating neurohumoral responses, activating growth factor cascades (such as TGF-β/Smad3 and PDGFs), inducing pro-inflammatory cytokines and chemokines, generating advanced glycation end-products (AGEs) and stimulating the AGE-RAGE axis, and upregulating fibrogenic matricellular proteins. Although diabetes-activated fibrogenic signaling has common characteristics in various tissues, some organs, such as the heart, kidney and liver develop more pronounced and clinically significant fibrosis. This review manuscript summarizes current knowledge on the cellular and molecular pathways involved in diabetic fibrosis, discussing the fundamental links between metabolic perturbations and fibrogenic activation, the basis for organ-specific differences, and the promises and challenges of anti-fibrotic therapies for diabetic patients.
Collapse
Affiliation(s)
- Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
3
|
Yang M, Ma X, Xuan X, Deng H, Chen Q, Yuan L. Liraglutide Attenuates Non-Alcoholic Fatty Liver Disease in Mice by Regulating the Local Renin-Angiotensin System. Front Pharmacol 2020; 11:432. [PMID: 32322207 PMCID: PMC7156971 DOI: 10.3389/fphar.2020.00432] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
The renin-angiotensin system (RAS) is involved in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) and represents a potential therapeutic target for NAFLD. Glucagon-like peptide-1 (GLP-1) signaling has been shown to regulate the RAS within various local tissues. In this study, we aimed to investigate the functional relationship between GLP-1 and the local RAS in the liver during NAFLD. Wild-type and ACE2 knockout mice were used to establish a high-fat-induced NAFLD model. After the mice were treated with liraglutide (a GLP-1 analogue) for 4 weeks, the key RAS component genes were up-regulated in the liver of NAFLD mice. Liraglutide treatment regulated the RAS balance, preventing a reduction in fatty acid oxidation gene expression and increasing gluconeogenesis and the expression of inflammation-related genes caused by NAFLD, which were impaired in ACE2 knockout mice. Liraglutide-treated HepG2 cells exhibited activation of the ACE2/Ang1-7/Mas axis, increased fatty acid oxidation gene expression, and decreased inflammation, which could be reversed by A779 and AngII. These results indicate that the local RAS in the liver becomes overactivated in response to NAFLD. Moreover, ACE2 knockout increases the severity of liver steatosis. Liraglutide has a negative and antagonistic effect on the ACE/AngII/AT1R axis, a positive impact on the ACE2/Ang1-7/Mas axis, and is mediated through the PI3K/AKT pathway. This may represent a potential new mechanism by which liraglutide improves NAFLD.
Collapse
Affiliation(s)
- Mengying Yang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyi Ma
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuping Xuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjun Deng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Reverte V, Gogulamudi VR, Rosales CB, Musial DC, Gonsalez SR, Parra-Vitela AJ, Galeas-Pena M, Sure VN, Visniauskas B, Lindsey SH, Katakam PVG, Prieto MC. Urinary angiotensinogen increases in the absence of overt renal injury in high fat diet-induced type 2 diabetic mice. J Diabetes Complications 2020; 34:107448. [PMID: 31761419 PMCID: PMC6981045 DOI: 10.1016/j.jdiacomp.2019.107448] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/09/2019] [Accepted: 09/07/2019] [Indexed: 12/31/2022]
Abstract
AIM OF THE STUDY During type 2 diabetes (T2D) and hypertension there is stimulation of renal proximal tubule angiotensinogen (AGT), but whether urinary excretion of AGT (uAGT) is an indicator of glomerular damage or intrarenal RAS activation is unclear. We tested the hypothesis that elevations in uAGT can be detected in the absence of albuminuria in a mouse model of T2D. METHODS Male C57BL/6 mice (N = 10) were fed a high fat (HFD; 45% Kcal from fat) for 28 weeks, and the metabolic phenotype including body weight, blood pressures, glucose, insulin, ippGTT, HOMA-IR, and cholesterol was examined. In addition, kidney Ang II content and reactive oxygen species (ROS) was measured along with urinary albumin, creatinine, Ang II, and AGT. RESULTS All parameters consistent with T2D were present in mice after 12-14 weeks on the HFD. Systolic BP increased after 18 weeks in HFD but not NFD mice. Intrarenal ROS and Ang II concentrations were also increased in HFD mice. Remarkably, these changes paralleled the augmentation uAGT excretion (3.66 ± 0.50 vs. 0.92 ± 0.13 ng/mg by week 29; P < 0.01), which occurred in the absence of overt albuminuria. CONCLUSIONS In HFD-induced T2D mice, increases in uAGT occur in the absence of overt renal injury, indicating that this biomarker accurately detects early intrarenal RAS activation.
Collapse
Affiliation(s)
- Virginia Reverte
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA
| | | | - Carla B Rosales
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA
| | - Diego C Musial
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA; Department of Pharmacology, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Sabrina R Gonsalez
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Michelle Galeas-Pena
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, USA
| | - Bruna Visniauskas
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, USA
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, USA
| | - Minolfa C Prieto
- Department of Physiology, Tulane University School of Medicine, New Orleans, USA; Hypertension and Renal Center of Excellence, New Orleans, USA.
| |
Collapse
|
5
|
Leite APO, Aragão DS, Nogueira MD, Pereira RO, Jara ZP, Fiorino P, Casarini DE, Farah V. Modulation of renin angiotensin system components by high glucose levels in the culture of collecting duct cells. J Cell Physiol 2019; 234:22809-22818. [DOI: 10.1002/jcp.28845] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 11/11/2022]
Affiliation(s)
- A. P. O. Leite
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| | - Danielle S. Aragão
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
| | - Marie D. Nogueira
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
| | - Renata O. Pereira
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| | - Zaira P. Jara
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Department of Molecular Cardiology Lerner Research Institute—Cleveland Clinic Cleveland Ohio
| | - Patricia Fiorino
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| | - Dulce E. Casarini
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
| | - Vera Farah
- Disciplina de Nefrologia, Departamento de Medicina, Escola Paulista de Medicina Universidade Federal de São Paulo São Paulo Brazil
- Laboratório de Renal, Cardiovascular e Fisiofarmacologia Metabólica, Centro de Ciência da Saúde e Biologia Universidade Presbiteriana Mackenzie São Paulo Brazil
| |
Collapse
|
6
|
Pahlavani M, Kalupahana NS, Ramalingam L, Moustaid-Moussa N. Regulation and Functions of the Renin-Angiotensin System in White and Brown Adipose Tissue. Compr Physiol 2017; 7:1137-1150. [PMID: 28915321 DOI: 10.1002/cphy.c160031] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The renin angiotensin system (RAS) is a major regulator of blood pressure, fluid, and electrolyte homeostasis. RAS precursor angiotensinogen (Agt) is cleaved into angiotensin I (Ang I) and II (Ang II) by renin and angiotensin converting enzyme (ACE), respectively. Major effects of Ang II, the main bioactive peptide of this system, is mediated by G protein coupled receptors, Angiotensin Type 1 (AGTR1, AT1R) and Type 2 (AGTR2, AT2R) receptors. Further, the discovery of additional RAS peptides such as Ang 1-7 generated by the action of another enzyme ACE2 identified novel functions of this complex system. In addition to the systemic RAS, several local RAS exist in organs such as the brain, kidney, pancreas, and adipose tissue. The expression and regulation of various components of RAS in adipose tissue prompted extensive research into the role of adipose RAS in metabolic diseases. Indeed, animal studies have shown that adipose-derived Agt contributes to circulating RAS, kidney, and blood pressure regulation. Further, mice overexpressing Agt have high blood pressure and increased adiposity characterized by inflammation, adipocyte hypertrophy, and insulin resistance, which can be reversed at least in part by RAS inhibition. These findings highlight the importance of this system in energy homeostasis, especially in the context of obesity. This overview article discusses the depot-specific functions of adipose RAS, genetic and pharmacological manipulations of RAS, and its applications to adipogenesis, thermogenesis, and overall energy homeostasis. © 2017 American Physiological Society. Compr Physiol 7:1137-1150, 2017.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| | - Nishan S Kalupahana
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA.,Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
7
|
Bruce EB, de Kloet AD. The intricacies of the renin-angiotensin-system in metabolic regulation. Physiol Behav 2017; 178:157-165. [PMID: 27887998 PMCID: PMC5600901 DOI: 10.1016/j.physbeh.2016.11.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 12/15/2022]
Abstract
Over recent years, the renin-angiotensin-system (RAS), which is best-known as an endocrine system with established roles in hydromineral balance and blood pressure control, has emerged as a fundamental regulator of many additional physiological and pathophysiological processes. In this manuscript, we celebrate and honor Randall Sakai's commitment to his trainees, as well as his contribution to science. Scientifically, Randall made many notable contributions to the recognition of the RAS's roles in brain and behavior. His interests, in this regard, ranged from its traditionally-accepted roles in hydromineral balance, to its less-appreciated functions in stress responses and energy metabolism. Here we review the current understanding of the role of the RAS in the regulation of metabolism. In particular, the opposing actions of the RAS within adipose tissue vs. its actions within the brain are discussed.
Collapse
Affiliation(s)
- Erin B Bruce
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, United States
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, United States.
| |
Collapse
|
8
|
The effects of apelin treatment on a rat model of type 2 diabetes. Adv Med Sci 2015; 60:94-100. [PMID: 25625368 DOI: 10.1016/j.advms.2014.11.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 11/17/2014] [Accepted: 11/25/2014] [Indexed: 02/08/2023]
Abstract
PURPOSE Apelin is an adipokine that plays a role in the regulation of many biological functions in mammals including the neuroendocrine, cardiovascular, immune systems, glucose homeostasis and obesity. It can act via autocrine, paracrine, endocrine, and exocrine signaling. We aimed to identify the role of apelin pathophysiology of diabetes. MATERIAL/METHODS 37 male Wistar Albino rats aged 8-10 weeks were divided in four experimental groups as: control group (C) control+apelin group (C+A), diabetic group (D) diabetic+apelin group (D+A). Apelin and apelin receptor mRNA gene expressions in heart and aorta tissue were determined by real-time polymerase chain reaction. The plasma levels of insulin and plasma apelin were determined by ELISA. RESULTS Plasma levels of insulin, glucose, blood pressure levels were significantly lower in D+A group. There was no statistically significant difference for level of apelin between diabetic groups. On the other hand, differences for apelin and APJ mRNA expression in heart and vascular tissue were found significant between groups. CONCLUSIONS Apelin can be used as a therapeutic agent in the treatment of type II diabetes in the future.
Collapse
|
9
|
Keshari KR, Wilson DM, Sai V, Bok R, Jen KY, Larson P, Van Criekinge M, Kurhanewicz J, Wang ZJ. Noninvasive in vivo imaging of diabetes-induced renal oxidative stress and response to therapy using hyperpolarized 13C dehydroascorbate magnetic resonance. Diabetes 2015; 64:344-52. [PMID: 25187363 PMCID: PMC4303960 DOI: 10.2337/db13-1829] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxidative stress has been proposed to be a unifying cause for diabetic nephropathy and a target for novel therapies. Here we apply a new endogenous reduction-oxidation (redox) sensor, hyperpolarized (HP) (13)C dehydroascorbate (DHA), in conjunction with MRI to noninvasively interrogate the renal redox capacity in a mouse diabetes model. The diabetic mice demonstrate an early decrease in renal redox capacity, as shown by the lower in vivo HP (13)C DHA reduction to the antioxidant vitamin C (VitC), prior to histological evidence of nephropathy. This correlates with lower tissue reduced glutathione (GSH) concentration and higher NADPH oxidase 4 (Nox4) expression, consistent with increased superoxide generation and oxidative stress. ACE inhibition restores the HP (13)C DHA reduction to VitC with concomitant normalization of GSH concentration and Nox4 expression in diabetic mice. HP (13)C DHA enables rapid in vivo assessment of altered redox capacity in diabetic renal injury and after successful treatment.
Collapse
Affiliation(s)
- Kayvan R Keshari
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David M Wilson
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Victor Sai
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Robert Bok
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Kuang-Yu Jen
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Peder Larson
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Mark Van Criekinge
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - John Kurhanewicz
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| | - Zhen J Wang
- Department of Radiology & Biomedical Imaging, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
10
|
Qi Y, Zhang K, Wu Y, Xu Z, Yong QC, Kumar R, Baker KM, Zhu Q, Chen S, Guo S. Novel mechanism of blood pressure regulation by forkhead box class O1-mediated transcriptional control of hepatic angiotensinogen. Hypertension 2014; 64:1131-40. [PMID: 25069665 DOI: 10.1161/hypertensionaha.114.03970] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The renin-angiotensin system is a major determinant of blood pressure regulation. It consists of a cascade of enzymatic reactions involving 3 components: angiotensinogen, renin, and angiotensin-converting enzyme, which generate angiotensin II as a biologically active product. Angiotensinogen is largely produced in the liver, acting as a major determinant of the circulating renin-angiotensin system, which exerts acute hemodynamic effects on blood pressure regulation. How the expression of angiotensinogen is regulated is not completely understood. Here, we hypothesize that angiotensinogen is regulated by forkhead transcription factor forkhead box class O1 (Foxo1), an insulin-suppressed transcription factor, and thereby controls blood pressure in mice. We generated liver-specific Foxo1 knockout mice, which exhibited a reduction in plasma angiotensinogen and angiotensin II levels and a significant decrease in blood pressure. Using hepatocyte cultures, we demonstrated that overexpression of Foxo1 increased angiotensinogen expression, whereas hepatocytes lacking Foxo1 demonstrated a reduction of angiotensinogen gene expression and partially impaired insulin inhibition on angiotensinogen gene expression. Furthermore, mouse angiotensinogen prompter analysis demonstrated that the angiotensinogen promoter region contains a functional Foxo1-binding site, which is responsible for both Foxo1 stimulation and insulin suppression on the promoter activity. Together, these data demonstrate that Foxo1 regulates hepatic angiotensinogen gene expression and controls plasma angiotensinogen and angiotensin II levels, modulating blood pressure control in mice.
Collapse
Affiliation(s)
- Yajuan Qi
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Kebin Zhang
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Yuxin Wu
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Zihui Xu
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Qian Chen Yong
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Rajesh Kumar
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Kenneth M Baker
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Qinglei Zhu
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Shouwen Chen
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.)
| | - Shaodong Guo
- From the Division of Molecular Cardiology, Department of Medicine, College of Medicine, Texas A&M University Health Science Center, Balyor Scott & White Health, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.); and Central Texas Veterans Health Care System, Temple (Y.Q., K.Z., Y.W., Z.X., Q.C.Y., R.K., K.M.B., Q.Z., S.C., S.G.).
| |
Collapse
|
11
|
Giese MJ, Speth RC. The ocular renin-angiotensin system: a therapeutic target for the treatment of ocular disease. Pharmacol Ther 2013; 142:11-32. [PMID: 24287313 DOI: 10.1016/j.pharmthera.2013.11.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 10/25/2013] [Indexed: 02/06/2023]
Abstract
The renin-angiotensin system (RAS) is most well-known for its role in regulation and dysregulation of blood pressure as well as fluid and electrolyte homeostasis. Due to its ability to cause cardiovascular disease, the RAS is the target of a multitude of drugs that antagonize its pathophysiological effects. While the "classical" RAS is a systemic hormonal system, there is an increasing awareness of the existence and functional significance of local RASs in a number of organs, e.g., liver, kidney, heart, lungs, reproductive organs, adipose tissue and adrenal. The eye is one of these organs where a compelling body of evidence has demonstrated the presence of a local RAS. Individual components of the RAS have been shown to be present in many structures of the eye and their potential functional significance in ocular disease states is described. Because the eye is one of the most important and complex organs in the body, this review also discusses the implications of dysregulation of the systemic RAS on the pathogenesis of ocular diseases and how pharmacological manipulation of the RAS might lead to novel or adjunctive therapies for ocular disease states.
Collapse
Affiliation(s)
| | - Robert C Speth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, United States.
| |
Collapse
|
12
|
Kopec AK, Boverhof DR, Nault R, Harkema JR, Tashiro C, Potter D, Sharratt B, Chittim B, Zacharewski TR. Toxicogenomic evaluation of long-term hepatic effects of TCDD in immature, ovariectomized C57BL/6 mice. Toxicol Sci 2013; 135:465-75. [PMID: 23864506 DOI: 10.1093/toxsci/kft156] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Acute exposure to hepatotoxic doses of 2,3,7,8-tetrachloro- dibenzo-p-dioxin (TCDD) in mice is characterized by differential gene expression that can be phenotypically anchored to elevated levels of serum alanine aminotransferase, increased relative liver weights, hepatic steatosis, inflammation, and hepatocellular necrosis. Unlike most studies that focus on acute exposure effects, this study evaluated the long-term effects of a single oral gavage of 30 μg/kg TCDD at 1, 4, 12, 24, 36, and 72 weeks postdose in ovariectomized C57BL/6 mice. Hepatic TCDD levels were almost completely eliminated by 24 weeks with a calculated half-life of 12 days. Hepatic gene expression analysis identified 395 unique differentially expressed genes between 1 and 12 weeks that decreased to ≤ 8 by 72 weeks, consistent with the minimal hepatic TCDD levels. Hepatic vacuolization, characteristic of short-term exposure, subsided by 4 weeks. Similarly, TCDD-elicited hepatic necrosis and inflammation dissipated by 1 week. Collectively, these results suggest that TCDD-elicited histologic and gene expression responses can be correlated to elevated hepatic TCDD levels, which, once eliminated, elicit minimal hepatic gene expression and histologic alterations.
Collapse
Affiliation(s)
- Anna K Kopec
- * Department of Biochemistry & Molecular Biology
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kalupahana NS, Moustaid-Moussa N. The renin-angiotensin system: a link between obesity, inflammation and insulin resistance. Obes Rev 2012; 13:136-49. [PMID: 22034852 DOI: 10.1111/j.1467-789x.2011.00942.x] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) is classically known for its role in regulation of blood pressure, fluid and electrolyte balance. Recently, several local RASs in organs such as brain, heart, pancreas and adipose tissue have also been identified. Evidence from clinical trials suggests that in addition to anti-hypertensive effects, pharmacological inhibition of RAS also provides protection against the development of type-2 diabetes. Moreover, animal models with targeted inactivation of RAS genes exhibit improved insulin sensitivity and are protected from high-fat diet-induced obesity and insulin resistance. Because there is evidence for RAS overactivation in obesity, it is possible that RAS is a link between obesity and insulin resistance. This review summarizes the evidence and mechanistic insights on the associations between RAS, obesity and insulin resistance, with special emphasis on the role of adipose tissue RAS in the pathogenesis of metabolic derangements in obesity.
Collapse
Affiliation(s)
- N S Kalupahana
- Obesity Research Center, The University of Tennessee, Knoxville, TN 37996-4588, USA
| | | |
Collapse
|
14
|
Putnam K, Shoemaker R, Yiannikouris F, Cassis LA. The renin-angiotensin system: a target of and contributor to dyslipidemias, altered glucose homeostasis, and hypertension of the metabolic syndrome. Am J Physiol Heart Circ Physiol 2012; 302:H1219-30. [PMID: 22227126 DOI: 10.1152/ajpheart.00796.2011] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The renin-angiotensin system (RAS) is an important therapeutic target in the treatment of hypertension. Obesity has emerged as a primary contributor to essential hypertension in the United States and clusters with other metabolic disorders (hyperglycemia, hypertension, high triglycerides, low HDL cholesterol) defined within the metabolic syndrome. In addition to hypertension, RAS blockade may also serve as an effective treatment strategy to control impaired glucose and insulin tolerance and dyslipidemias in patients with the metabolic syndrome. Hyperglycemia, insulin resistance, and/or specific cholesterol metabolites have been demonstrated to activate components required for the synthesis [angiotensinogen, renin, angiotensin-converting enzyme (ACE)], degradation (ACE2), or responsiveness (angiotensin II type 1 receptors, Mas receptors) to angiotensin peptides in cell types (e.g., pancreatic islet cells, adipocytes, macrophages) that mediate specific disorders of the metabolic syndrome. An activated local RAS in these cell types may contribute to dysregulated function by promoting oxidative stress, apoptosis, and inflammation. This review will discuss data demonstrating the regulation of components of the RAS by cholesterol and its metabolites, glucose, and/or insulin in cell types implicated in disorders of the metabolic syndrome. In addition, we discuss data supporting a role for an activated local RAS in dyslipidemias and glucose intolerance/insulin resistance and the development of hypertension in the metabolic syndrome. Identification of an activated RAS as a common thread contributing to several disorders of the metabolic syndrome makes the use of angiotensin receptor blockers and ACE inhibitors an intriguing and novel option for multisymptom treatment.
Collapse
Affiliation(s)
- Kelly Putnam
- Graduate Center for Nutritional Sciences, University of Kentucky, Lexington, 40536-0200, USA
| | | | | | | |
Collapse
|
15
|
Influence of obesity and metabolic dysfunction on the endothelial control in the coronary circulation. J Mol Cell Cardiol 2011; 52:840-7. [PMID: 21889942 DOI: 10.1016/j.yjmcc.2011.08.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 08/17/2011] [Accepted: 08/18/2011] [Indexed: 02/07/2023]
Abstract
Diseases of the coronary circulation remain the leading cause of death in Western society despite impressive advances in diagnosis, pharmacotherapy and post-event management. Part of this statistic likely stems from a parallel increase in the prevalence of obesity and metabolic dysfunction, both significant risk factors for coronary disease. Obesity and diabetes pose unique challenges for the heart and their impact on the coronary vasculature remains incompletely understood. The vascular endothelium is a major interface between arterial function and the physical and chemical components of blood flow. Proper function of the endothelium is necessary to preserve hemostasis, maintain vascular tone and limit the extent of vascular diseases such as atherosclerosis. Given its central role in vascular health, endothelial dysfunction has been the source of considerable research interest in diabetes and obesity. In the current review, we will examine the pathologic impact of obesity and diabetes on coronary function and the extent to which these two factors impact endothelial function. This article is part of a Special Issue entitled "Coronary Blood Flow".
Collapse
|
16
|
Giannini C, Mohn A, Chiarelli F, Kelnar CJH. Macrovascular angiopathy in children and adolescents with type 1 diabetes. Diabetes Metab Res Rev 2011; 27:436-60. [PMID: 21433262 DOI: 10.1002/dmrr.1195] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Diabetes represents one of the most common diseases globally. Worryingly, the worldwide incidence of type 1 diabetes (T1D) is rising by 3% per year. Despite the rapid increase in diabetes incidence, recent advances in diabetes treatment have been successful in decreasing morbidity and mortality from diabetes-related retinopathy, nephropathy, and neuropathy. In contrast, there is clear evidence for the lack of improvement in mortality for cardiovascular diseases (CVDs). This emphasizes the importance of focusing childhood diabetes care strategies for the prevention of CVD in adulthood. Furthermore, although most work on diabetes and macrovascular disease relates to type 2 diabetes, it has been shown that the age-adjusted relative risk of CVD in T1D far exceeds that in type 2 diabetes. As T1D appears predominantly during childhood, those with T1D are at greater risk for coronary events early in life and require lifelong medical attention. Because of the important health effects of CVDs in children and adolescents with T1D, patients, family members, and care providers should understand the interaction of T1D and cardiovascular risk. In addition, optimal cardiac care for the patient with diabetes should focus on aggressive management of traditional cardiovascular risk factors to optimize those well-recognized as well as new specific risk factors which are becoming available. Therefore, a complete characterization of the molecular mechanisms involved in the development and progression of macrovascular angiopathy is needed. Furthermore, as vascular abnormalities begin as early as in childhood, potentially modifiable risk factors should be identified at an early stage of vascular disease development.
Collapse
Affiliation(s)
- Cosimo Giannini
- Department of Pediatrics, University of Chieti, Chieti, Italy.
| | | | | | | |
Collapse
|
17
|
Carvalho CR, Bueno AA, Mattos AM, Biz C, de Oliveira C, Pisani LP, Ribeiro EB, Oller do Nascimento CM, Oyama LM. Fructose alters adiponectin, haptoglobin and angiotensinogen gene expression in 3T3-L1 adipocytes. Nutr Res 2010; 30:644-9. [DOI: 10.1016/j.nutres.2010.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 06/04/2010] [Accepted: 06/17/2010] [Indexed: 11/30/2022]
|
18
|
de Kloet AD, Krause EG, Woods SC. The renin angiotensin system and the metabolic syndrome. Physiol Behav 2010; 100:525-34. [PMID: 20381510 PMCID: PMC2886177 DOI: 10.1016/j.physbeh.2010.03.018] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 03/18/2010] [Accepted: 03/26/2010] [Indexed: 01/01/2023]
Abstract
The renin angiotensin system (RAS; most well-known for its critical roles in the regulation of cardiovascular function and hydromineral balance) has regained the spotlight for its potential roles in various aspects of the metabolic syndrome. It may serve as a causal link among obesity and several co-morbidities. Drugs that reduce the synthesis or action of angiotensin-II (A-II; the primary effector peptide of the RAS) have been used to treat hypertension for decades and, more recently, clinical trials have determined the utility of these pharmacological agents to prevent insulin resistance. Moreover, there is evidence that the RAS contributes to body weight regulation by acting in various tissues. This review summarizes what is known of the actions of the RAS in the brain and throughout the body to influence various metabolic disorders. Special emphasis is given to the role of the RAS in body weight regulation. The paper represents an invited review by a symposium, award winner or keynote speaker at the Society for the Study of Ingestive Behavior [SSIB] Annual Meeting in Portland, July 2009.
Collapse
Affiliation(s)
- Annette D de Kloet
- Program in Neuroscience University of Cincinnati, Cincinnati, OH 45237, United States.
| | | | | |
Collapse
|
19
|
Santos SHS, Braga JF, Mario EG, Pôrto LCJ, Rodrigues-Machado MDG, Murari A, Botion LM, Alenina N, Bader M, Santos RAS. Improved lipid and glucose metabolism in transgenic rats with increased circulating angiotensin-(1-7). Arterioscler Thromb Vasc Biol 2010; 30:953-61. [PMID: 20203301 DOI: 10.1161/atvbaha.109.200493] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Obesity and diabetes remain among the world's most pervasive health problems. Although the importance of angiotensin II for metabolic regulation is well documented, the role of the angiotensin-(1-7)/Mas axis in this process is poorly understood. The aim of this study was to evaluate the effect of increased angiotensin-(1-7) plasma levels in lipid and glucose metabolism using transgenic rats that express an angiotensin-(1-7)-releasing fusion protein, TGR(A1-7)3292 (TGR). METHODS AND RESULTS The increased angiotensin-(1-7) levels in TGR induced enhanced glucose tolerance, insulin sensitivity, and insulin-stimulated glucose uptake. In addition, TGR presented decreased triglycerides and cholesterol levels, as well as a significant decrease in abdominal fat mass, despite normal food intake. These alterations were accompanied by a marked decrease of angiotensinogen expression and increased Akt in adipose tissue. Furthermore, augmented plasma levels and expression in adipose tissue was observed for adiponectin. Accordingly, angiotensin-(1-7) stimulation increased adiponectin production by primary adipocyte culture, which was blocked by the Mas antagonist A779. Circulating insulin and muscle glycogen content were not altered in TGR. CONCLUSION These results show that increased circulating angiotensin-(1-7) levels lead to prominent changes in glucose and lipid metabolism.
Collapse
|
20
|
Steckelings UM, Rompe F, Kaschina E, Unger T. The evolving story of the RAAS in hypertension, diabetes and CV disease - moving from macrovascular to microvascular targets. Fundam Clin Pharmacol 2009; 23:693-703. [DOI: 10.1111/j.1472-8206.2009.00780.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
21
|
Siddique RA, Tandon M, Ambwani T, Rai SN, Atreja SK. Nutrigenomics: Nutrient-Gene Interactions. FOOD REVIEWS INTERNATIONAL 2009. [DOI: 10.1080/87559120903155883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
22
|
TRC4186, a Novel AGE-breaker, Improves Diabetic Cardiomyopathy and Nephropathy in Ob-ZSF1 Model of Type 2 Diabetes. J Cardiovasc Pharmacol 2009; 54:72-81. [DOI: 10.1097/fjc.0b013e3181ac3a34] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Laczy B, Hill BG, Wang K, Paterson AJ, White CR, Xing D, Chen YF, Darley-Usmar V, Oparil S, Chatham JC. Protein O-GlcNAcylation: a new signaling paradigm for the cardiovascular system. Am J Physiol Heart Circ Physiol 2009; 296:H13-28. [PMID: 19028792 PMCID: PMC2637779 DOI: 10.1152/ajpheart.01056.2008] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Accepted: 11/11/2008] [Indexed: 02/07/2023]
Abstract
The posttranslational modification of serine and threonine residues of nuclear and cytoplasmic proteins by the O-linked attachment of the monosaccharide beta-N-acetylglucosamine (O-GlcNAc) is a highly dynamic and ubiquitous protein modification. Protein O-GlcNAcylation is rapidly emerging as a key regulator of critical biological processes including nuclear transport, translation and transcription, signal transduction, cytoskeletal reorganization, proteasomal degradation, and apoptosis. Increased levels of O-GlcNAc have been implicated as a pathogenic contributor to glucose toxicity and insulin resistance, which are both major hallmarks of diabetes mellitus and diabetes-related cardiovascular complications. Conversely, there is a growing body of data demonstrating that the acute activation of O-GlcNAc levels is an endogenous stress response designed to enhance cell survival. Reports on the effect of altered O-GlcNAc levels on the heart and cardiovascular system have been growing rapidly over the past few years and have implicated a role for O-GlcNAc in contributing to the adverse effects of diabetes on cardiovascular function as well as mediating the response to ischemic injury. Here, we summarize our present understanding of protein O-GlcNAcylation and its effect on the regulation of cardiovascular function. We examine the pathways regulating protein O-GlcNAcylation and discuss, in more detail, our understanding of the role of O-GlcNAc in both mediating the adverse effects of diabetes as well as its role in mediating cellular protective mechanisms in the cardiovascular system. In addition, we also explore the parallels between O-GlcNAc signaling and redox signaling, as an alternative paradigm for understanding the role of O-GlcNAcylation in regulating cell function.
Collapse
Affiliation(s)
- Boglarka Laczy
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294-0007, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Genetic variation in the renin-angiotensin system modifies the beneficial effects of ACE inhibitors on the risk of diabetes mellitus among hypertensives. J Hum Hypertens 2008; 22:774-80. [PMID: 18563171 DOI: 10.1038/jhh.2008.62] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The aim of this study was to assess whether the association between angiotensin-converting enzyme (ACE) inhibitor use and the incidence of treated diabetes mellitus is modified by genetic polymorphisms in the renin-angiotensin system (RAS).In a nested case-control study, treated hypertensive patients were genotyped for ACE (insertion (I)/deletion (D)), angiotensinogen (AGT; M235T) and angiotensin II type 1 receptor (AGTR1; A1166C). Cases of newly treated diabetes were identified based on pharmacy records and controls were not yet drug treated for diabetes (case:control ratio 1:10). Self-administered questionnaires and physical examinations were used to assess risk factors for diabetes mellitus. Logistic regression was used to calculate the relative risk of diabetes associated with ACE inhibitor use relative to other antihypertensive treatment, stratified by the RAS genotypes. Among 205 cases and 2050 controls, homozygous 1166A carriers of the AGTR1 gene had a significantly decreased incidence of diabetes associated with current use of ACE inhibitors (odds ratio, OR: 0.47; 95% CI: 0.26-0.84), whereas this incidence was increased among 1166C allele carriers (OR: 1.32; 95% CI: 0.81-2.14). The interaction OR was 3.21 (95% CI: 1.53-6.75). ACE I allele carriers had a significantly reduced incidence of diabetes associated with ACE inhibitors use (OR: 0.63; 95% CI: 0.41-0.98), whereas DD homozygotes had no reduced risk (OR: 0.95; 95% CI: 0.46-1.96). The risk of diabetes associated with ACE inhibitor use was not significantly modified by the AGT-M235T polymorphism. Treatment with ACE inhibitors in hypertensive subjects significantly reduces the occurrence of diabetes in homozygous 1166A carriers of the AGTR1 gene and carriers of the ACE I allele, but not in 1166C allele carriers of the AGTR1 gene and in homozygous ACE D allele carriers.
Collapse
|
25
|
Genetically altered animals in the study of the metabolic functions of peptide hormone systems. Curr Opin Nephrol Hypertens 2008; 17:11-7. [PMID: 18090664 DOI: 10.1097/mnh.0b013e3282f2909a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Here we review the use of genetically altered animals to address the roles of peptide hormone systems in the modulation of energy homeostasis. Despite the disseminated use of transgenic techniques to establish the functional relevance of several peptide hormone systems, we focus on two multifunctional systems, the renin-angiotensin and the kallikrein-kinin systems. Initially, we explored the background information supporting the functional aspects of these systems, followed by novel knowledge obtained with the phenotypic characterization of genetically altered animals. RECENT FINDINGS A role for the renin-angiotensin system in the regulation of adiposity and glucose metabolism has been suggested. Studies using genetically altered animals not only confirmed the physiological relevance of angiotensin II in the control of energy homeostasis, but also revealed that the adipose tissue renin-angiotensin system participates in the endocrine modulation of cardiovascular and renal function. On the other hand, the involvement of the kallikrein-kinin system with metabolic processes was not so obvious. Recent reports using genetically altered animals, however, provided strong evidence to support an important role for kinins in the control of glucose homeostasis and energy balance. SUMMARY Here we present examples of how genetically altered animals contribute to a final postulation of the physiological roles of certain hormone systems, bringing new insights into the field.
Collapse
|
26
|
Santos SHS, Fernandes LR, Mario EG, Ferreira AVM, Pôrto LCJ, Alvarez-Leite JI, Botion LM, Bader M, Alenina N, Santos RAS. Mas deficiency in FVB/N mice produces marked changes in lipid and glycemic metabolism. Diabetes 2008; 57:340-7. [PMID: 18025412 DOI: 10.2337/db07-0953] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Metabolic syndrome is characterized by the variable coexistence of obesity, hyperinsulinemia, insulin resistance, dyslipidemia, and hypertension. It is well known that angiotensin (Ang) II is importantly involved in the metabolic syndrome. However, the role of the vasodilator Ang-(1-7)/Mas axis is not known. The aim of this study was to evaluate the effect of genetic deletion of the G protein-coupled receptor, Mas, in the lipidic and glycemic metabolism in FVB/N mice. RESEARCH DESIGN AND METHODS Plasma lipid, insulin, and cytokine concentrations were measured in FVB/N Mas-deficient and wild-type mice. A glucose tolerance test was performed by intraperitoneally injecting d-glucose into overnight-fasted mice. An insulin sensitivity test was performed by intraperitoneal injection of insulin. Uptake of 2-deoxy-[(3)H]glucose by adipocytes was used to determine the rate of glucose transport; adipose tissue GLUT4 was quantified by Western blot. Gene expression of transforming growth factor (TGF)-beta, type 1 Ang II receptor, and angiotensinogen (AGT) were measured by real-time PCR. RESULTS Despite normal body weight, Mas-knockout (Mas-KO) mice presented dyslipidemia, increased levels of insulin and leptin, and an approximately 50% increase in abdominal fat mass. In addition, Mas gene-deleted mice presented glucose intolerance and reduced insulin sensitivity as well as a decrease in insulin-stimulated glucose uptake by adipocytes and decreased GLUT4 in adipose tissue. Mas(-/-) presented increased muscle triglycerides, while liver triglyceride levels were normal. Expression of TGF-beta and AGT genes was higher in Mas-KO animals in comparison with controls. CONCLUSIONS These results show that Mas deficiency in FVB/N mice leads to dramatic changes in glucose and lipid metabolisms, inducing a metabolic syndrome-like state.
Collapse
Affiliation(s)
- Sérgio Henrique S Santos
- Laborátorio de Hipertensão, Departamento de Fisiologia e Biofisica, Universidade Federal de Minas Gerais, Av Antonio Carlos 6627-ICB, 31270-901, Belo Horizonte, Minas Gerais, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Takeshita Y, Takamura T, Ando H, Hamaguchi E, Takazakura A, Matsuzawa-Nagata N, Kaneko S. Cross talk of tumor necrosis factor-α and the renin–angiotensin system in tumor necrosis factor-α-induced plasminogen activator inhibitor-1 production from hepatocytes. Eur J Pharmacol 2008; 579:426-32. [DOI: 10.1016/j.ejphar.2007.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Revised: 11/02/2007] [Accepted: 11/14/2007] [Indexed: 10/22/2022]
|
28
|
Jansson L, Andersson A, Bodin B, Källskog O. Pancreatic islet blood flow during euglycaemic, hyperinsulinaemic clamp in anaesthetized rats. Acta Physiol (Oxf) 2007; 189:319-24. [PMID: 17367401 DOI: 10.1111/j.1748-1716.2006.01666.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
AIMS Previous studies have demonstrated that pancreatic islet blood flow is crucially dependent on blood glucose concentration. Thus, hyperglycaemia increases and hypoglycaemia decreases islet blood perfusion, by a combination of nervous and metabolic signals. The aim of the present study was to evaluate if hyperinsulinaemia, without associated hypoglycaemia, affects islet blood flow. METHODS Thiobutabarbital-anaesthetized Wistar-Furth rats were subjected to an euglycaemic, hyperinsulinaemic clamp, that is they were infused for 60 min with either saline, insulin (18 mU kg(-1) min(-1)), glucose (27 mg kg(-1) min(-1)) or both glucose and insulin. This was followed by islet blood flow measurements with a microsphere technique. RESULTS Animals receiving only glucose doubled their blood glucose and serum insulin concentrations, whereas rats receiving only insulin had blood glucose concentrations <2 mmol L(-1) and a 10-fold increase in serum insulin concentrations. Animals given simultaneous glucose and insulin had normal blood glucose concentrations but a 10-fold increase in serum insulin concentrations. Total pancreatic blood flow was unaffected in all animals. Islet blood flow was increased in hyperglycaemic and decreased in hypoglycaemic rats compared with control rats. Islet blood flow did not differ between clamped and control rats. CONCLUSIONS Serum insulin concentration per se does not affect islet blood flow, whereas the ambient blood glucose concentration is of major importance in this context.
Collapse
Affiliation(s)
- L Jansson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
29
|
Fülöp N, Marchase RB, Chatham JC. Role of protein O-linked N-acetyl-glucosamine in mediating cell function and survival in the cardiovascular system. Cardiovasc Res 2007; 73:288-97. [PMID: 16970929 PMCID: PMC2848961 DOI: 10.1016/j.cardiores.2006.07.018] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2006] [Revised: 07/14/2006] [Accepted: 07/18/2006] [Indexed: 10/24/2022] Open
Abstract
There is growing recognition that the O-linked attachment of N-acetyl-glucosamine (O-GlcNAc) on serine and threonine residues of nuclear and cytoplasmic proteins is a highly dynamic post-translational modification that plays a key role in signal transduction pathways. Numerous proteins have been identified as targets of O-GlcNAc modifications including kinases, phosphatases, transcription factors, metabolic enzymes, chaperons, and cytoskeletal proteins. Modulation of O-GlcNAc levels has been shown to modify DNA binding, enzyme activity, protein-protein interactions, the half-life of proteins, and subcellular localization. The level of O-GlcNAc is regulated in part by the metabolism of glucose via the hexosamine biosynthesis pathway (HBP), and the metabolic abnormalities associated with insulin resistance and diabetes, such as hyperglycemia, hyperlipidemia, and hyperinsulinemia, are all associated with increased flux through the HBP and elevated O-GlcNAc levels. Increased HBP flux and O-GlcNAc levels have been implicated in the impaired relaxation of isolated cardiomyocytes, blunted response to angiotensin II and phenylephrine, hyperglycemia-induced cardiomyocyte apoptosis, and endothelial and vascular cell dysfunction. In contrast to these adverse effects, recent studies have also shown that O-GlcNAc levels increase in response to acute stress and that this is associated with increased cell survival. Thus, while the relationship between O-GlcNAc levels and cellular function is complex and not well-understood, it is clear that these pathways play a critical role in the regulation of cell function and survival in the cardiovascular system and may be implicated in the adverse effects of metabolic disease on the heart.
Collapse
Affiliation(s)
- Norbert Fülöp
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - Richard B. Marchase
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - John C. Chatham
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW In diabetes, oxidative stress plays a key role in the pathogenesis of vascular complications; therefore an antioxidant therapy would be of great interest in this disease. RECENT FINDINGS Hyperglycemia directly promotes an endothelial dysfunction--inducing process of overproduction of superoxide at the mitochondrial level. This is the first and key event able to activate all the pathways involved in the development of vascular complications of diabetes. It has recently been shown that statins, angiotensin-converting enzyme inhibitors, angiotensin II type 1 blockers, calcium channel blockers, and thiazolidinediones have a strong intracellular antioxidant activity. SUMMARY Classic antioxidants, such as vitamin E, failed to show beneficial effects on diabetic complications probably because their action is only "symptomatic". The preventive activity against hyperglycemia-induced oxidative stress shown by statins, angiotensin-converting enzyme inhibitors, angiotensin II type 1 blockers, calcium channel blockers, and thiazolidinediones justifies use of these compounds for preventing complications in patients with diabetes, in whom antioxidant defences have been shown to be defective.
Collapse
Affiliation(s)
- Antonio Ceriello
- Warwick Medical School, Clinical Science Research Institute, University Hospital-Walsgrave Campus, University of Warwick, Clifford Bridge Road, Coventry CV2 2DX, UK.
| |
Collapse
|
31
|
Cheng DW, Jiang Y, Shalev A, Kowluru R, Crook ED, Singh LP. An analysis of high glucose and glucosamine-induced gene expression and oxidative stress in renal mesangial cells. Arch Physiol Biochem 2006; 112:189-218. [PMID: 17178593 DOI: 10.1080/13813450601093518] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Renal mesangial cells play an important role in the development of diabetic kidney disease. We have previously demonstrated that some of the effects of high glucose on mesangial extracellular matrix (ECM) protein expression are mediated by the hexosamine biosynthesis pathway (HBP) in which fructose-6-phosphate is converted to glucosamine-6-phosphate by the rate-limiting enzyme glutamine:fructose-6-phosphate amidotransferase (GFAT). Using Affymetrix murine expression U430 2.0 oligochips, we examined the global effects of high glucose (HG) and glucosamine (GlcN) on mRNA expression of a mouse mesangial cell line (MES-13). We sought to determine the portion of mRNA expression in MES-13 cells, which is mediated both by high glucose and glucosamine, i.e., via the HBP. Of the 34,000 genes on the chip, approximately 55.7 - 60.8% genes are detected in MES-13 cells. Culturing MES-13 cells for 48 h with HG alters the expression of approximately 389 genes at our preset threshold levels (at least 2-fold change) where 263 genes are up-regulated and 126 genes are down-regulated. GlcN also increases the expression of 106 genes and decreases 94 genes during the same period of incubation. Seventy-two genes in the chip are commonly regulated by HG and GlcN, in which 33 genes are up and 39 genes are down. The mRNA level of thioredoxin interacting protein (TXNIP), an inhibitor of thioredoxin activity, is maximally increased approximately 18.8 and 9.9-fold respectively by HG and GlcN. The differential expression of several genes found in the microarray analysis is further validated by real-time quantitative PCR. Significant biological processes commonly targeted by HG and GlcN are the TXNIP-thioredoxin system, oxidative stress, endoplasmic reticulum (ER) stress, extracellular matrix genes, and interferon-inducible genes. Stable overexpression of TXNIP in MES-13 cells increases glucose and glucosamine-mediated ECM gene expression and oxidative stress. We conclude from these results that the HBP mediates several effects of high glucose on mesangial cell metabolism, which promotes reactive oxygen species generation to cause cellular oxidative stress, ECM gene expression and apoptosis.
Collapse
Affiliation(s)
- Davis W Cheng
- Department of Anatomy, Wayne State University School of Medicine, MI 48201, USA
| | | | | | | | | | | |
Collapse
|
32
|
Kim MJ, Lee DH, Park DB, Kang HW, An CS, Cui XJ, Kang JS, Kim JL, Lee YJ, Jung KW. Chronic blockade of the angiotensin II receptor has a differential effect on adipose and vascular PAI-1 in OLETF rats. Diabetes Res Clin Pract 2006; 73:8-16. [PMID: 16413628 DOI: 10.1016/j.diabres.2005.11.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2005] [Revised: 11/08/2005] [Accepted: 11/25/2005] [Indexed: 10/25/2022]
Abstract
Angiotensinogen (AGT) and plasminogen activator inhibitor-1 (PAI-1) are expressed in both vascular and adipose tissues. Angiotensin II (AG II) has an adipogenic effect and increases PAI-1 expression. To evaluate the chronic effects of AG II type 1 receptor (AT(1)R) antagonism on adipose mass and PAI-1 expression in vascular and adipose tissues, losartan (30mg/kg/day) was administered to Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a model of type 2 diabetes, for 20 weeks. Adipose mass and regional fat distribution in the abdomen did not change after chronic AT(1)R antagonism in OLETF rats. AGT and PAI-1 mRNA expressions in adipose tissue of OLETF rats were significantly increased compared with Long-Evans Tokushima Otsuka (LETO) rats, the normal control. Chronic losartan therapy further increased the level of adipose AGT in OLETF rats, but did not affect the level of adipose PAI-1 mRNA. In contrast, aortic PAI-1 expression in OLETF rats was attenuated by chronic losartan therapy. Our results have two implications. First, adipose tissue may be an important source of AG II in metabolic syndrome even after chronic losartan therapy. Second, chronic AT(1)R antagonism with losartan causes differential effects on vascular and adipose PAI-1 expression.
Collapse
Affiliation(s)
- Myeung-Ju Kim
- Department of Anatomy, College of Medicine, Dankook University, San 29, Anseo-dong, Cheonan-si, Chung-nam 330-714, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Thomas MC, Tikellis C, Burns WM, Bialkowski K, Cao Z, Coughlan MT, Jandeleit-Dahm K, Cooper ME, Forbes JM. Interactions between Renin Angiotensin System and Advanced Glycation in the Kidney. J Am Soc Nephrol 2005; 16:2976-84. [PMID: 16107577 DOI: 10.1681/asn.2005010013] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Although hemodynamic and metabolic factors are individually implicated in the development of diabetic nephropathy, their interaction has not been defined clearly. In this study, the effects of angiotensin II (Ang II) and advanced glycation end products (AGE) both individually on each other are explored and compared. In the first study arm, Sprague-Dawley rats received a continuous infusion of AGE-modified rat serum albumin (RSA) or unmodified RSA for 4 wk with or without the angiotensin receptor type 1 antagonist valsartan. In the second arm, animals received a continuous infusion of Ang II (58.3 ng/kg per min) with or without the AGE inhibitor pyridoxamine. Components of the intrarenal renin-angiotensin system were measured using real time reverse transcription-PCR, immunohistochemistry, and standard angiotensin-converting enzyme (ACE) activity assays. Renal and serum AGE were quantified by immunohistochemistry, ELISA, and AGE-fluorescence. After an infusion of AGE-RSA, renal expression of angiotensinogen, ACE, renin, and angiotensin receptor type 1 were increased significantly (all P < 0.01), and ACE activity was elevated. This was associated with tubular and glomerular hypertrophy and AGE accumulation, which could be antagonized by valsartan. However, valsartan had no effect on increased filtration fraction associated with an AGE-RSA infusion. At the same time, an infusion of Ang II increased the serum and renal accumulation of AGE and advanced oxidation protein products and induced renal hypertrophy and salt retention that could be antagonized by pyridoxamine. However, pyridoxamine had no effect on renal vasoconstriction manifested by reduced renal blood flow. AGE and Ang II have overlapping activities in the kidney. The beneficial effects of blockade of either pathway underline the importance of this interaction in diabetic renal disease and the aging kidney.
Collapse
Affiliation(s)
- Merlin C Thomas
- Baker Medical Research Institute, P.O. Box 6492, Melbourne, Victoria 8008, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Chiarelli F, Di Marzio D, Santilli F, Mohn A, Blasetti A, Cipollone F, Mezzetti A, Verrotti A. Effects of irbesartan on intracellular antioxidant enzyme expression and activity in adolescents and young adults with early diabetic angiopathy. Diabetes Care 2005; 28:1690-7. [PMID: 15983321 DOI: 10.2337/diacare.28.7.1690] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Defective intracellular antioxidant enzyme production (IAP) has been demonstrated in adults with diabetic nephropathy. The objective of this study was to evaluate the effects of irbesartan, an angiotensin II receptor antagonist, on IAP in adolescents and young adults with type 1 diabetes and early signs of retinopathy and nephropathy. RESEARCH DESIGN AND METHODS This prospective, matched case-control study was conducted between November 2001 and December 2002 among 14 type 1 diabetic patients with early signs of angiopathy (ages 14-21 years), 11 type 1 diabetic patients without angiopathy (ages 12-22 years), and 10 healthy volunteers (ages 16-22 years). Skin fibroblasts were obtained by skin biopsies from the anterior part of the forearm and cultured in Dulbecco's modified Eagle's medium. The activity and mRNA expression of CuZn superoxide dismutase (CuZnSOD), Mn superoxide dismutase (MnSOD), catalase (CAT), and glutathione peroxidase (GPX) were measured before and after 6 months of treatment with irbesartan (150 mg/day); on both occasions, antioxidant enzyme activity was evaluated at different glucose concentrations (5 and 22 mmol/l). RESULTS At a normal glucose concentration (5 mmol/l), the activity and mRNA expression of CuZnSOD (0.50 +/- 0.21 units/mg protein, 4.4 +/- 1.5 mRNA/glyceraldehyde-3-phosphate dehydrogenase), MnSOD (0.26 +/- 0.04 units/mg protein, 0.08 +/- 0.07 mRNA), CAT (0.32 +/- 0.08 units/mg protein, 4.8 +/- 1.3 mRNA), and GPX (0.53 +/- 0.09 units/mg protein, 2.2 +/- 0.9 mRNA) were not different among the three groups (only values of diabetic subjects with angiopathy are shown). At high glucose concentrations, the activity and mRNA expression of CuZnSOD increased similarly in all groups (diabetic subjects with angiopathy: 0.93 +/- 0.26 units/mg protein, 9.4 +/- 2.1 mRNA); that of CAT and GPX increased in only control subjects and diabetic subjects without angiopathy (diabetic subjects with angiopathy: 0.33 +/- 0.09 units/mg protein and 5.0 +/- 1.4 mRNA; 0.54 +/- 0.10 units/mg protein and 2.3 +/- 1.0 mRNA, respectively). MnSOD did not change in any group. Treatment with irbesartan in adolescents with diabetic angiopathy was able to restore CAT and GPX activity and mRNA expression after exposure to high glucose concentrations. Markers of oxidative stress (serum malondialdehyde, fluorescent products of lipid peroxidation, monocyte chemoattractant protein-1, and 8-isoprostanes prostaglandin F(2alpha)) were significantly reduced after treatment with irbesartan. CONCLUSIONS Adolescents and young adults with early signs of diabetic angiopathy have defective intracellular antioxidant enzyme production and activity. Treatment with irbesartan can substantially improve the activity and production of these enzymes in skin fibroblasts.
Collapse
Affiliation(s)
- Francesco Chiarelli
- Department of Pediatrics, University of Chieti, Ospedale Policlinico, Via dei Vestini, 5, I-66100 Chieti, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Einstein FH, Atzmon G, Yang XM, Ma XH, Rincon M, Rudin E, Muzumdar R, Barzilai N. Differential responses of visceral and subcutaneous fat depots to nutrients. Diabetes 2005; 54:672-8. [PMID: 15734842 DOI: 10.2337/diabetes.54.3.672] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Increased visceral adiposity is a pivotal component of the metabolic syndrome. Differential gene expression patterns of fat-derived peptides (FDPs) in visceral fat and subcutaneous fat have been characterized in the fasting state. Here we examined whether delivery of nutrients differentially affects the expression of FDPs in visceral fat versus subcutaneous fat (in the fed state). We increased the rate of glucose flux into adipose tissue of normal rats (n = 16) by hyperglycemia or hyperinsulinemia using the clamp technique. Glucose uptake was associated with increased expression of FDPs, including resistin ( approximately 5-fold), adiponectin ( approximately 2-fold), leptin ( approximately 15-fold), plasminogen activating inhibitor-1 ( approximately 10-fold), and angiotensinogen ( approximately 4-fold) in visceral fat, but markedly less in subcutaneous fat. Cytokine expression derived mainly from vascular/stromal/macrophage components of adipose tissue was less dramatically increased. Infusion of glucosamine amplified the results obtained by increasing glucose uptake into adipose tissue, suggesting that flux through the hexosamine biosynthetic pathway may serve as a mechanism for "nutrient sensing." Nutrient-dependent expression of FDPs in visceral fat was also associated with increased plasma levels of several FDPs. Because a biologic sensing pathway can dynamically couple daily food intake to abnormal plasma levels of important FDPs, we challenge the practice of obtaining plasma levels after fasting to assess risk factors for metabolic syndrome.
Collapse
Affiliation(s)
- Francine H Einstein
- Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Laverty S, Sandy JD, Celeste C, Vachon P, Marier JF, Plaas AHK. Synovial fluid levels and serum pharmacokinetics in a large animal model following treatment with oral glucosamine at clinically relevant doses. ACTA ACUST UNITED AC 2005; 52:181-91. [PMID: 15641100 DOI: 10.1002/art.20762] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To examine the concentration of glucosamine in the synovial fluid and its pharmacokinetics in serum in a large animal model following dosing with glucosamine HCl at clinically relevant levels. METHODS Eight adult female horses were studied. After an overnight fast, glucosamine HCl (20 mg/kg of body weight) was administered by either nasogastric (NG) intubation or intravenous (IV) injection. Blood samples were collected before dosing and at 5, 15, 30, 60, 120, 180, 240, 360, 480, and 720 minutes after dosing. Synovial fluid samples were collected from the radiocarpal joints 48 hours before dosing and at 1 and 12 hours after dosing. Glucosamine was assayed by fluorophore-assisted carbohydrate electrophoresis. RESULTS The maximum concentration of glucosamine in serum reached approximately 300 muM ( approximately 50 microg/ml) following IV dosing and approximately 6 microM (approximately 1 microg/ml) following NG dosing. Synovial fluid concentrations reached 9-15 microM with IV dosing and 0.3-0.7 microM with NG dosing, and remained elevated (range 0.1-0.7 microM) in most animals even at 12 hours after dosing. Following NG dosing, the median serum maximal concentration of 6.1 microM (range 4.38-7.58) was attained between 30 minutes and 4 hours postdose. The mean apparent volume of distribution was 15.4 liters/kg, the mean bioavailability was 5.9%, and the mean elimination half-life was 2.82 hours. CONCLUSION Clinically relevant dosing of glucosamine HCl in this large monogastric animal model results in serum and synovial fluid concentrations that are at least 500-fold lower than those reported to modify chondrocyte anabolic and catabolic activities in tissue and cell culture experiments. We conclude that the apparent therapeutic benefit of dietary glucosamine on pain and joint space width in humans and animals may be secondary to its effects on nonarticular tissues, such as the intestinal lining, liver, or kidney, since these may be exposed to much high levels of glucosamine following ingestion.
Collapse
Affiliation(s)
- Sheila Laverty
- Faculté de Médicine Vétérinaire, Université de Montréal, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
37
|
Atwood CS, Barzilai N, Bowen RL, Brown-Borg HM, Jarrard DF, Fu VX, Heilbronn LK, Ingram DK, Ravussin E, Schwartz RS, Weindruch R. Pennington scientific symposium on mechanisms and retardation of aging. Exp Gerontol 2004; 38:1217-26. [PMID: 14580876 DOI: 10.1016/j.exger.2003.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Craig S Atwood
- Section of Geriatrics and Gerontology, University of Wisconsin Medical School, Madison, WI, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Tan Y, Hutchison FN, Jaffa AA. Mechanisms of angiotensin II-induced expression of B2 kinin receptors. Am J Physiol Heart Circ Physiol 2004; 286:H926-32. [PMID: 14766673 DOI: 10.1152/ajpheart.00757.2003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although the primary roles of the kallikreinkinin system and the renin-angiotensin system are quite divergent, they are often intertwined under pathophysiological conditions. We examined the effect of ANG II on regulation of B(2) kinin receptors (B2KR) in vascular cells. Vascular smooth muscle cells (VSMC) were treated with ANG II in a concentration (10(-9)-10(-6) M)- and time (0-24 h)-dependent manner, and B2KR protein and mRNA levels were measured by Western blots and PCR, respectively. A threefold increase in B2KR protein levels was observed as early as 6 h, with a peak response at 10(-7) M. ANG II (10(-7) M) also increased B2KR mRNA levels twofold 4 h after stimulation. Actinomycin D suppressed the increase in B2KR mRNA and protein levels induced by ANG II. To elucidate the receptor subtype involved in mediating this regulation, VSMC were pretreated with losartan (AT(1) receptor antagonist) and/or PD-123319 (AT(2) receptor antagonist) at 10 microM for 30 min, followed by ANG II (10(-7) M) stimulation. Losartan completely blocked the ANG II-induced B2KR increase, whereas PD-123319 had no effect. In addition, expression of B2KR mRNA levels was decreased in AT(1A) receptor knockout mice. Finally, to determine whether ANG II stimulates B2KR expression via activation of the MAPK pathway, VSMC were pretreated with an inhibitor of p42/p44(mapk) (PD-98059) and/or an inhibitor of p38(mapk) (SB-202190), followed by ANG II (10(-7) M) for 24 h. Selective inhibition of the p42/p44(mapk) pathway significantly blocked the ANG II-induced increase in B2KR expression. These findings demonstrate that ANG II regulates expression of B2KR in VSMC and provide a rationale for studying the interaction between ANG II and bradykinin in the pathogenesis of vascular dysfunction.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Aorta/cytology
- Cells, Cultured
- Extracellular Fluid/metabolism
- Gene Expression/drug effects
- Male
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/physiology
- Rats
- Rats, Sprague-Dawley
- Receptor, Bradykinin B2/genetics
- Receptor, Bradykinin B2/metabolism
- Signal Transduction/drug effects
- Vasoconstrictor Agents/pharmacology
Collapse
Affiliation(s)
- Yan Tan
- Dept. of Medicine, Endocrinology-Diabetes-Medical Genetics, Medical Univ. of South Carolina, 114 Doughty St., PO Box 250776, Charleston, SC 29425, USA
| | | | | |
Collapse
|
39
|
Schmaier AH. The kallikrein-kinin and the renin-angiotensin systems have a multilayered interaction. Am J Physiol Regul Integr Comp Physiol 2003; 285:R1-13. [PMID: 12793984 DOI: 10.1152/ajpregu.00535.2002] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Understanding the physiological role of the plasma kallikrein-kinin system (KKS) has been hampered by not knowing how the proteins of this proteolytic system, when assembled in the intravascular compartment, become activated under physiological conditions. Recent studies indicate that the enzyme prolylcarboxypeptidase, an ANG II inactivating enzyme, is a prekallikrein activator. The ability of prolylcarboxypeptidase to act in the KKS and the renin-angiotensin system (RAS) indicates a novel interaction between these two systems. This interaction, along with the roles of angiotensin converting enzyme, cross talk between bradykinin and angiotensin-(1-7) action, and the opposite effects of activation of the ANG II receptors 1 and 2 support a hypothesis that the plasma KKS counterbalances the RAS. This review examines the interaction and cross talk between these two protein systems. This analysis suggests that there is a multilayered interaction between these two systems that are important for a wide array of physiological functions.
Collapse
Affiliation(s)
- Alvin H Schmaier
- The Univ. of Michigan, 5301 MSRB III, 1150 West Medical Center Dr., Ann Arbor, MI 48109-0640, USA.
| |
Collapse
|
40
|
Abstract
Many studies have demonstrated that excess of visceral fat has deleterious effects on insulin action. Mainly, it has been shown to be associated with a decrease in hepatic and peripheral insulin sensitivity, which results in a clinical condition also known as insulin resistance. This report describes a novel experimental method that we employed in order to analyze the particular effects of visceral fat on insulin activity. By extracting visceral fat we were able to distinguish the specific role that it plays in insulin action, and to analyze its effects on the gene expression of a variety of fat-derived peptides, which may be considered to be (at least partially) mediators in the development of the metabolic syndrome and possibly diabetes mellitus.
Collapse
Affiliation(s)
- Ilan Gabriely
- Institute for Aging Research, Belfer Building #701, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | |
Collapse
|
41
|
Ceriello A. New insights on oxidative stress and diabetic complications may lead to a "causal" antioxidant therapy. Diabetes Care 2003; 26:1589-96. [PMID: 12716823 DOI: 10.2337/diacare.26.5.1589] [Citation(s) in RCA: 502] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Evidence implicates hyperglycemia-derived oxygen free radicals as mediators of diabetic complications. However, intervention studies with classic antioxidants, such as vitamin E, failed to demonstrate any beneficial effect. Recent studies demonstrate that a single hyperglycemia-induced process of overproduction of superoxide by the mitochondrial electron-transport chain seems to be the first and key event in the activation of all other pathways involved in the pathogenesis of diabetic complications. These include increased polyol pathway flux, increased advanced glycosylation end product formation, activation of protein kinase C, and increased hexosamine pathway flux. Superoxide overproduction is accompanied by increased nitric oxide generation, due to an endothelial NOS and inducible NOS uncoupled state, a phenomenon favoring the formation of the strong oxidant peroxynitrite, which in turn damages DNA. DNA damage is an obligatory stimulus for the activation of the nuclear enzyme poly(ADP-ribose) polymerase. Poly(ADP-ribose) polymerase activation in turn depletes the intracellular concentration of its substrate NAD(+), slowing the rate of glycolysis, electron transport, and ATP formation, and produces an ADP-ribosylation of the GAPDH. These processes result in acute endothelial dysfunction in diabetic blood vessels that, convincingly, also contributes to the development of diabetic complications. These new findings may explain why classic antioxidants, such as vitamin E, which work by scavenging already-formed toxic oxidation products, have failed to show beneficial effects on diabetic complications and may suggest new and attractive "causal" antioxidant therapy. New low-molecular mass compounds that act as SOD or catalase mimetics or L-propionyl-carnitine and lipoic acid, which work as intracellular superoxide scavengers, improving mitochondrial function and reducing DNA damage, may be good candidates for such a strategy, and preliminary studies support this hypothesis. This "causal" therapy would also be associated with other promising tools such as LY 333531, PJ34, and FP15, which block the protein kinase beta isoform, poly(ADP-ribose) polymerase, and peroxynitrite, respectively. While waiting for these focused tools, we may have other options: thiazolinediones, statins, ACE inhibitors, and angiotensin 1 inhibitors can reduce intracellular oxidative stress generation, and it has been suggested that many of their beneficial effects, even in diabetic patients, are due to this property.
Collapse
Affiliation(s)
- Antonio Ceriello
- Department of Pathology and Medicine, Experimental and Clinical, University of Udine, Italy.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Angiotensinogen and components of the renin-angiotensin system are expressed in adipose tissue of rodents and humans, but the role of generated angiotensin II has remained intriguing. Moreover, the functional importance of angiotensin II receptor subtypes in preadipocytes and adipocytes still remains a controversial subject. RECENT FINDINGS Recent findings in transgenic mice have emphasized the upregulation of angiotensinogen expression by glucocorticoids. Furthermore, angiotensinogen products, that is angiotensin II and possibly angiotensin II-related products, have been found to act locally in modulating adipose tissue growth in an autocrine/paracrine manner. Cellularity measurements show that fat mass enlargement is associated with adipocyte hypertrophy, consistent with the upregulation of the fatty acid synthetase gene by angiotensin II depicted at the molecular level. Together, these findings suggest a mechanism by which transient or chronic overexpression of angiotensinogen in adipose tissue favors lipogenesis in adipocytes and leads to a 'vicious' circle whereby adipose tissue development is further increased. SUMMARY Additional studies are warranted to characterize angiotensin II-related receptors, if any, and to clarify the role played by angiotensin II receptor subtypes and metabolites in various metabolic aspects of white adipose tissue.
Collapse
Affiliation(s)
- Gérard Ailhaud
- Institute of Signaling, Developmental Biology and Cancer Research, Centre de Biochimie (UMR 6543 CNRS), Laboratory Biology of Adipose Tissue Development, Faculté des Sciences, Parc Valrose, Nice, France.
| | | | | |
Collapse
|