1
|
Alves-Silva T, Húngaro TG, Freitas-Lima LC, de Melo Arthur G, Arruda AC, Santos RB, Oyama LM, Mori MA, Bader M, Araujo RC. Kinin B1 receptor controls maternal adiponectin levels and influences offspring weight gain. iScience 2023; 26:108409. [PMID: 38058311 PMCID: PMC10696114 DOI: 10.1016/j.isci.2023.108409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/16/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023] Open
Abstract
Given the importance of the kinin B1 receptor in insulin and leptin hormonal regulation, which in turn is crucial in maternal adaptations to ensure nutrient supply to the fetus, we investigated the role of this receptor in maternal metabolism and fetoplacental development. Wild-type and kinin B1 receptor-deficient (B1KO) female mice were mated with male mice of the opposite genotype. Consequently, the entire litter was heterozygous for kinin B1 receptor, ensuring that there would be no influence of offspring genotype on the maternal phenotype. Maternal kinin B1 receptor blockade reduces adiponectin secretion by adipose tissue ex vivo, consistent with lower adiponectin levels in pregnant B1KO mice. Furthermore, fasting insulinemia also increased, which was associated with placental insulin resistance, reduced placental glycogen accumulation, and heavier offspring. Therefore, we propose the combination of chronic hyperinsulinemia and reduced adiponectin secretion in B1KO female mice create a maternal obesogenic environment that results in heavier pups.
Collapse
Affiliation(s)
- Thaís Alves-Silva
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
- Max-Delbrück Center for Molecular Medicine (MDC), Campus Berlin-Buch, 13125 Berlin, Germany
| | - Talita G.R. Húngaro
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Leandro C. Freitas-Lima
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Gabriel de Melo Arthur
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Adriano C. Arruda
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Raisa B. Santos
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| | - Lila M. Oyama
- Laboratory of Nutrition and Endocrine Physiology, Physiology Department, Federal University of São Paulo (UNIFESP), São Paulo 04023-901, Brazil
| | - Marcelo A.S. Mori
- Laboratory of Aging Biology, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo 13083-862, Brazil
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine (MDC), Campus Berlin-Buch, 13125 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
- Charité University Medicine Berlin, Berlin, Germany
| | - Ronaldo C. Araujo
- Laboratory of Genetics and Exercise Metabolism, Molecular Biology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
- Laboratory of Genetics and Exercise Metabolism, Nephrology Program, Biophysics Department, Federal University of São Paulo (UNIFESP), São Paulo 04039-032, Brazil
| |
Collapse
|
2
|
Cannabinoids Transmogrify Cancer Metabolic Phenotype via Epigenetic Reprogramming and a Novel CBD Biased G Protein-Coupled Receptor Signaling Platform. Cancers (Basel) 2023; 15:cancers15041030. [PMID: 36831374 PMCID: PMC9954791 DOI: 10.3390/cancers15041030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/29/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
The concept of epigenetic reprogramming predicts long-term functional health effects. This reprogramming can be activated by exogenous or endogenous insults, leading to altered healthy and different disease states. The exogenous or endogenous changes that involve developing a roadmap of epigenetic networking, such as drug components on epigenetic imprinting and restoring epigenome patterns laid down during embryonic development, are paramount to establishing youthful cell type and health. This epigenetic landscape is considered one of the hallmarks of cancer. The initiation and progression of cancer are considered to involve epigenetic abnormalities and genetic alterations. Cancer epigenetics have shown extensive reprogramming of every component of the epigenetic machinery in cancer development, including DNA methylation, histone modifications, nucleosome positioning, non-coding RNAs, and microRNA expression. Endocannabinoids are natural lipid molecules whose levels are regulated by specific biosynthetic and degradative enzymes. They bind to and activate two primary cannabinoid receptors, type 1 (CB1) and type 2 (CB2), and together with their metabolizing enzymes, form the endocannabinoid system. This review focuses on the role of cannabinoid receptors CB1 and CB2 signaling in activating numerous receptor tyrosine kinases and Toll-like receptors in the induction of epigenetic landscape alterations in cancer cells, which might transmogrify cancer metabolism and epigenetic reprogramming to a metastatic phenotype. Strategies applied from conception could represent an innovative epigenetic target for preventing and treating human cancer. Here, we describe novel cannabinoid-biased G protein-coupled receptor signaling platforms (GPCR), highlighting putative future perspectives in this field.
Collapse
|
3
|
Rouhiainen A, Kulesskaya N, Mennesson M, Misiewicz Z, Sipilä T, Sokolowska E, Trontti K, Urpa L, McEntegart W, Saarnio S, Hyytiä P, Hovatta I. The bradykinin system in stress and anxiety in humans and mice. Sci Rep 2019; 9:19437. [PMID: 31857655 PMCID: PMC6923437 DOI: 10.1038/s41598-019-55947-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 11/27/2019] [Indexed: 01/06/2023] Open
Abstract
Pharmacological research in mice and human genetic analyses suggest that the kallikrein-kinin system (KKS) may regulate anxiety. We examined the role of the KKS in anxiety and stress in both species. In human genetic association analysis, variants in genes for the bradykinin precursor (KNG1) and the bradykinin receptors (BDKRB1 and BDKRB2) were associated with anxiety disorders (p < 0.05). In mice, however, neither acute nor chronic stress affected B1 receptor gene or protein expression, and B1 receptor antagonists had no effect on anxiety tests measuring approach-avoidance conflict. We thus focused on the B2 receptor and found that mice injected with the B2 antagonist WIN 64338 had lowered levels of a physiological anxiety measure, the stress-induced hyperthermia (SIH), vs controls. In the brown adipose tissue, a major thermoregulator, WIN 64338 increased expression of the mitochondrial regulator Pgc1a and the bradykinin precursor gene Kng2 was upregulated after cold stress. Our data suggests that the bradykinin system modulates a variety of stress responses through B2 receptor-mediated effects, but systemic antagonists of the B2 receptor were not anxiolytic in mice. Genetic variants in the bradykinin receptor genes may predispose to anxiety disorders in humans by affecting their function.
Collapse
Affiliation(s)
- Ari Rouhiainen
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Natalia Kulesskaya
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Marie Mennesson
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland.,SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Neuroscience Center, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Zuzanna Misiewicz
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland
| | - Tessa Sipilä
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Ewa Sokolowska
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Kalevi Trontti
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland.,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland.,SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Neuroscience Center, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lea Urpa
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - William McEntegart
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Suvi Saarnio
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland
| | - Petri Hyytiä
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland
| | - Iiris Hovatta
- Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland. .,Department of Psychology and Logopedics, Medicum, University of Helsinki, Helsinki, Finland. .,SleepWell Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Neuroscience Center, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| |
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: 10] [Impact Index Per Article: 2.0] [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
|
Jayathilake C, Visvanathan R, Deen A, Bangamuwage R, Jayawardana BC, Nammi S, Liyanage R. Cowpea: an overview on its nutritional facts and health benefits. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:4793-4806. [PMID: 29656381 DOI: 10.1002/jsfa.9074] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 02/16/2018] [Accepted: 04/04/2018] [Indexed: 05/23/2023]
Abstract
Cowpea (Vigna unguiculata) is a legume consumed as a high-quality plant protein source in many parts of the world. High protein and carbohydrate contents with a relatively low fat content and a complementary amino acid pattern to that of cereal grains make cowpea an important nutritional food in the human diet. Cowpea has gained more attention recently from consumers and researchers worldwide as a result of its exerted health beneficial properties, including anti-diabetic, anti-cancer, anti-hyperlipidemic, anti-inflammatory and anti-hypertensive properties. Among the mechanisms that have been proposed in the prevention of chronic diseases, the most proven are attributed to the presence of compounds such as soluble and insoluble dietary fiber, phytochemicals, and proteins and peptides in cowpea. However, studies on the anti-cancer and anti-inflammatory properties of cowpea have produced conflicting results. Some studies support a protective effect of cowpea on the progression of cancer and inflammation, whereas others did not reveal any. Because there are only a few studies addressing health-related effects of cowpea consumption, further studies in this area are suggested. In addition, despite the reported favorable effects of cowpea on diabetes, hyperlipidemia and hypertension, a long-term epidemiological study investigating the association between cowpea consumption and diabetes, cardiovascular disease and cancer is also recommended. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chathuni Jayathilake
- Division of Nutritional Biochemistry, National Institute of Fundamental Studies, Kandy, Sri Lanka
| | - Rizliya Visvanathan
- Division of Nutritional Biochemistry, National Institute of Fundamental Studies, Kandy, Sri Lanka
| | - Afka Deen
- Division of Nutritional Biochemistry, National Institute of Fundamental Studies, Kandy, Sri Lanka
| | - Ruksheela Bangamuwage
- Division of Nutritional Biochemistry, National Institute of Fundamental Studies, Kandy, Sri Lanka
| | | | - Srinivas Nammi
- School of Science and Health, Western Sydney University, Sydney, NSW, Australia
- National Institute of Complementary Medicine (NICM), Western Sydney University, Sydney, NSW, Australia
| | - Ruvini Liyanage
- Division of Nutritional Biochemistry, National Institute of Fundamental Studies, Kandy, Sri Lanka
| |
Collapse
|
6
|
Biased G protein-coupled receptor agonism mediates Neu1 sialidase and matrix metalloproteinase-9 crosstalk to induce transactivation of insulin receptor signaling. Cell Signal 2018; 43:71-84. [DOI: 10.1016/j.cellsig.2017.12.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/26/2017] [Accepted: 12/21/2017] [Indexed: 11/19/2022]
|
7
|
Frigolet ME, Thomas G, Beard K, Lu H, Liu L, Fantus IG. The bradykinin-cGMP-PKG pathway augments insulin sensitivity via upregulation of MAPK phosphatase-5 and inhibition of JNK. Am J Physiol Endocrinol Metab 2017; 313:E321-E334. [PMID: 28679626 DOI: 10.1152/ajpendo.00298.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/13/2023]
Abstract
Bradykinin (BK) promotes insulin sensitivity and glucose uptake in adipocytes and other cell types. We demonstrated that in rat adipocytes BK enhances insulin-stimulated glucose transport via endothelial nitric oxide synthase, nitric oxide (NO) generation, and decreased activity of the mitogen-activated protein kinase (MAPK) JNK (c-Jun NH2-terminal kinase). In endothelial cells, NO increases soluble guanylate cyclase (sGC) activity, which, in turn, activates protein kinase G (PKG) by increasing cGMP levels. In this study, we investigated whether BK acts via the sGC-cGMP-PKG pathway to inhibit the negative effects of JNK on insulin signaling and glucose uptake in rat adipocytes. BK augmented cGMP concentrations. The BK-induced enhancement of insulin-stimulated glucose uptake was mimicked by the sGC activator YC-1 and a cell-permeable cGMP analog, CPT-cGMP, and inhibited by the sGC inhibitor ODQ and the PKG inhibitor KT 5823. Transfection of dominant-negative PKG reduced the BK augmentation of insulin-induced Akt phosphorylation. The activation of JNK and ERK1/2 by insulin was attenuated by BK, which was mediated by the sGC-cGMP-PKG pathway. Whereas insulin-stimulated phosphorylation of upstream activators of JNK and ERK, i.e., MKK4 and MEK1/2, was unaffected, BK augmented insulin-mediated induction of MKP-5 mRNA and protein levels. Furthermore, zaprinast, a phosphodiesterase inhibitor, enhanced cGMP and MKP-5 and prolonged the action of BK. These data indicate that BK enhances insulin action by inhibition of negative feedback by JNK and ERK via upregulation of MKP-5, mediated by the sGC-cGMP-PKG signaling pathway.
Collapse
Affiliation(s)
- María E Frigolet
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
| | - Garry Thomas
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kristin Beard
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Huogen Lu
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lijiang Liu
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - I George Fantus
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada;
- Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, Toronto, Ontario, Canada; and
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Morais RL, Silva ED, Sales VM, Filippelli-Silva R, Mori MA, Bader M, Pesquero JB. Kinin B1 and B2 receptor deficiency protects against obesity induced by a high-fat diet and improves glucose tolerance in mice. Diabetes Metab Syndr Obes 2015; 8:399-407. [PMID: 26346752 PMCID: PMC4554409 DOI: 10.2147/dmso.s87635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The kallikrein-kinin system is well known for its role in pain and inflammation, and has been shown recently by our group to have a role also in the regulation of energy expenditure. We have demonstrated that B1 receptor knockout (B1KO) mice are resistant to obesity induced by a high-fat diet (HFD) and that B1 receptor expression in adipocytes regulates glucose tolerance and predisposition to obesity. However, it is also known that in the absence of B1 receptor, the B2 receptor is overexpressed and can take over the function of its B1 counterpart, rendering uncertain the role of each kinin receptor in these metabolic effects. Therefore, we investigated the impact of ablation of each kinin receptor on energy metabolism using double kinin receptor knockout (B1B2KO) mice. Our data show that B1B2KO mice were resistant to HFD-induced obesity, with lower food intake and feed efficiency when compared with wild-type mice. They also had lower blood insulin and leptin levels and higher glucose tolerance after treatment with an HFD. Gene expression for tumor necrosis factor-alpha and C-reactive protein, which are important genes for insulin resistance, was reduced in white adipose tissue, skeletal muscle, and the liver in B1B2KO mice after the HFD. In summary, our data show that disruption of kinin B1 and B2 receptors has a profound impact on metabolic homeostasis in mice, by improving glucose tolerance and preventing HFD-induced obesity. These novel findings could pave the way for development of new pharmacological strategies to treat metabolic disorders such as insulin resistance and obesity.
Collapse
Affiliation(s)
- Rafael L Morais
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Elton D Silva
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vicência M Sales
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Marcelo A Mori
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - João B Pesquero
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
- Correspondence: João B Pesquero, Department of Biophysics, Universidade Federal de São Paulo, Pedro de Toledo, 669 - 9th floor, Vila Clementino 04039-034, São Paulo, SP, Brazil, Tel +55 11 5576 4848 ext 1214, Email
| |
Collapse
|
9
|
Kolodka T, Charles ML, Raghavan A, Radichev IA, Amatya C, Ellefson J, Savinov AY, Nag A, Williams MS, Robbins MS. Preclinical characterization of recombinant human tissue kallikrein-1 as a novel treatment for type 2 diabetes mellitus. PLoS One 2014; 9:e103981. [PMID: 25100328 PMCID: PMC4123992 DOI: 10.1371/journal.pone.0103981] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 07/04/2014] [Indexed: 01/06/2023] Open
Abstract
Modulation of the kallikrein-kinin system (KKS) has been shown to have beneficial effects on glucose homeostasis and several other physiological responses relevant to the progression of type 2 diabetes mellitus (T2D). The importance of bradykinin and its receptors in mediating these responses is well documented, but the role of tissue kallikrein-1, the protease that generates bradykinin insitu, is much less understood. We developed and tested DM199, recombinant human tissue kallikrein-1 protein (rhKLK-1), as a potential novel therapeutic for T2D. Hyperinsulinemic-euglycemic clamp studies suggest that DM199 increases whole body glucose disposal in non-diabetic rats. Single-dose administration of DM199 in obese db/db mice and ZDF rats, showed an acute, dose-dependent improvement in whole-body glucose utilization. Sub-acute dosing for a week in ZDF rats improved glucose utilization, with a concomitant rise in fasting insulin levels and HOMA1-%B scores. After cessation of sub-acute dosing, fasting blood glucose levels were significantly lower in ZDF rats during a drug wash-out period. Our studies show for the first time that DM199 administration results in acute anti-hyperglycemic effects in several preclinical models, and demonstrate the potential for further development of DM199 as a novel therapeutic for T2D.
Collapse
Affiliation(s)
- Tadeusz Kolodka
- DiaMedica USA, Inc., Minneapolis, Minnesota, United States of America
| | | | - Arvind Raghavan
- DiaMedica USA, Inc., Minneapolis, Minnesota, United States of America
- * E-mail:
| | - Ilian A. Radichev
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Christina Amatya
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Jacob Ellefson
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Alexei Y. Savinov
- Sanford Project/Children’s Health Research Center, Sanford Research, Sioux Falls, South Dakota, United States of America
| | - Abhijeet Nag
- Invitek, Inc., Hayward, California, United States of America
| | - Mark S. Williams
- DiaMedica USA, Inc., Minneapolis, Minnesota, United States of America
| | - Mark S. Robbins
- DiaMedica USA, Inc., Minneapolis, Minnesota, United States of America
| |
Collapse
|
10
|
Potier L, Waeckel L, Fumeron F, Bodin S, Fysekidis M, Chollet C, Bellili N, Bonnet F, Gusto G, Velho G, Marre M, Alhenc-Gelas F, Roussel R, Bouby N. Tissue kallikrein deficiency, insulin resistance, and diabetes in mouse and man. J Endocrinol 2014; 221:297-308. [PMID: 24599937 DOI: 10.1530/joe-13-0529] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The kallikrein-kinin system has been suggested to participate in the control of glucose metabolism. Its role and the role of angiotensin-I-converting enzyme, a major kinin-inactivating enzyme, are however the subject of debate. We have evaluated the consequence of deficiency in tissue kallikrein (TK), the main kinin-forming enzyme, on the development of insulin resistance and diabetes in mice and man. Mice with inactivation of the TK gene were fed a high-fat diet (HFD) for 3 months, or crossed with obese, leptin-deficient (ob/ob) mice to generate double ob/ob-TK-deficient mutants. In man, a loss-of-function polymorphism of the TK gene (R53H) was studied in a large general population cohort tested for insulin resistance, the DESIR study (4843 participants, 9 year follow-up). Mice deficient in TK gained less weight on the HFD than their WT littermates. Fasting glucose level was increased and responses to glucose (GTT) and insulin (ITT) tolerance tests were altered at 10 and 16 weeks on the HFD compared with standard on the diet, but TK deficiency had no influence on these parameters. Likewise, ob-TK⁻/⁻ mice had similar GTT and ITT responses to those of ob-TK⁺/⁺ mice. TK deficiency had no effect on blood pressure in either model. In humans, changes over time in BMI, fasting plasma glucose, insulinemia, and blood pressure were not influenced by the defective 53H-coding TK allele. The incidence of diabetes was not influenced by this allele. These data do not support a role for the TK-kinin system, protective or deleterious, in the development of insulin resistance and diabetes.
Collapse
Affiliation(s)
- Louis Potier
- INSERM U1138, Centre de Recherche des Cordeliers, 15 rue de l'Ecole de Médecine, 75006 Paris, France University Paris Descartes, Paris, France University Pierre and Marie Curie, Paris, France Diabetology, Endocrinology and Nutrition Department, AP-HP, Bichat-Claude Bernard Hospital, Paris, France INSERM U695, Paris, France Paris Diderot University, Paris 7, Paris, France Department of Endocrinology, CHU Rennes, Univ Rennes 1, Inserm UMR 991, Rennes, France Institut inter Régional Pour la Santé, Irsa, La Riche, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Ahimastos AA, Latouche C, Natoli AK, Reddy-luthmoodoo M, Golledge J, Kingwell BA. Potential Vascular Mechanisms of Ramipril Induced Increases in Walking Ability in Patients With Intermittent Claudication. Circ Res 2014; 114:1144-55. [DOI: 10.1161/circresaha.114.302420] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
We recently reported that ramipril more than doubled maximum walking times in patients with peripheral artery disease with intermittent claudication.
Objective:
Our aim was to conduct exploratory analyses of the effects of ramipril therapy on circulating biomarkers of angiogenesis/arteriogenesis, thrombosis, inflammation, and leukocyte adhesion in patients with intermittent claudication.
Methods and Results:
One hundred sixty-five patients with intermittent claudication (mean, 65.3 [SD, 6.7] years) were administered ramipril 10 mg per day (n=82) or matching placebo (n=83) for 24 weeks in a randomized, double-blind study. Plasma biomarkers of angiogenesis/arteriogenesis (vascular endothelial growth factor-A, fibroblast growth factor-2), thrombosis (D-dimer, von Willebrand factor, thrombin-antithrombin III), inflammation (high-sensitivity C-reactive protein, osteopontin), and leukocyte adhesion (soluble vascular cell adhesion molecule-1, soluble intracellular adhesion molecule-1) were measured at baseline and 24 weeks. Relative to placebo, ramipril was associated with increases in vascular endothelial growth factor-A by 38% (95% confidence interval [CI], 34%–42%) and fibroblast growth factor-2 by 64% (95% CI, 44–85%;
P
<0.001 for both), and reductions in D-dimer by 24% (95% CI, −30% to −18%), von Willebrand factor by 22% (95% CI, −35% to −9%), thrombin-antithrombin III by 16% (95% CI, −19% to −13%), high-sensitivity C-reactive protein by 13% (95% CI, −14% to −9%), osteopontin by 12% (95% CI, −14% to −10%), soluble vascular cell adhesion molecule-1 by 14% (95% CI, −18% to −10%), and soluble intracellular adhesion molecule-1 by 15% (95% CI, −17% to −13%; all
P
<0.001). With the exception of von Willebrand factor, all the above changes correlated significantly with the change in maximum walking time (
P
=0.02−0.001) in the group treated with ramipril.
Conclusions:
Ramipril is associated with an increase in the biomarkers of angiogenesis/arteriogenesis and reduction in the markers of thrombosis, inflammation, and leukocyte adhesion. This study informs strategies to improve mobility in patients with intermittent claudication.
Clinical Trial Registration Information:
URL:
http://clinicaltrials.gov
. Unique identifier: NCT00681226.
Collapse
Affiliation(s)
- Anna A. Ahimastos
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Celine Latouche
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Alaina K. Natoli
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Medini Reddy-luthmoodoo
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Jonathan Golledge
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| | - Bronwyn A. Kingwell
- From Baker IDI Heart and Diabetes Institute and Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia (A.A.A., C.L., A.K.N., M.R., B.A.K.); Queensland Research Centre for Peripheral Vascular Disease, James Cook University, Townsville, Queensland, Australia (J.G.); and Department of Vascular and Endovascular Surgery, Townsville Hospital, Queensland, Australia (J.G.)
| |
Collapse
|
12
|
Non-canonical signalling and roles of the vasoactive peptides angiotensins and kinins. Clin Sci (Lond) 2014; 126:753-74. [DOI: 10.1042/cs20130414] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
GPCRs (G-protein-coupled receptors) are among the most important targets for drug discovery due to their ubiquitous expression and participation in cellular events under both healthy and disease conditions. These receptors can be activated by a plethora of ligands, such as ions, odorants, small ligands and peptides, including angiotensins and kinins, which are vasoactive peptides that are classically involved in the pathophysiology of cardiovascular events. These peptides and their corresponding GPCRs have been reported to play roles in other systems and under pathophysiological conditions, such as cancer, central nervous system disorders, metabolic dysfunction and bone resorption. More recently, new mechanisms have been described for the functional regulation of GPCRs, including the transactivation of other signal transduction receptors and the activation of G-protein-independent pathways. The existence of such alternative mechanisms for signal transduction and the discovery of agonists that can preferentially trigger one signalling pathway over other pathways (called biased agonists) have opened new perspectives for the discovery and development of drugs with a higher specificity of action and, therefore, fewer side effects. The present review summarizes the current knowledge on the non-canonical signalling and roles of angiotensins and kinins.
Collapse
|
13
|
The renin-angiotensin system in adipose tissue and its metabolic consequences during obesity. J Nutr Biochem 2013; 24:2003-15. [PMID: 24120291 DOI: 10.1016/j.jnutbio.2013.07.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/24/2013] [Accepted: 07/22/2013] [Indexed: 02/07/2023]
Abstract
Obesity is a worldwide disease that is accompanied by several metabolic abnormalities such as hypertension, hyperglycemia and dyslipidemia. The accelerated adipose tissue growth and fat cell hypertrophy during the onset of obesity precedes adipocyte dysfunction. One of the features of adipocyte dysfunction is dysregulated adipokine secretion, which leads to an imbalance of pro-inflammatory, pro-atherogenic versus anti-inflammatory, insulin-sensitizing adipokines. The production of renin-angiotensin system (RAS) components by adipocytes is exacerbated during obesity, contributing to the systemic RAS and its consequences. Increased adipose tissue RAS has been described in various models of diet-induced obesity (DIO) including fructose and high-fat feeding. Up-regulation of the adipose RAS by DIO promotes inflammation, lipogenesis and reactive oxygen species generation and impairs insulin signaling, all of which worsen the adipose environment. Consequently, the increase of circulating RAS, for which adipose tissue is partially responsible, represents a link between hypertension, insulin resistance in diabetes and inflammation during obesity. However, other nutrients and food components such as soy protein attenuate adipose RAS, decrease adiposity, and improve adipocyte functionality. Here, we review the molecular mechanisms by which adipose RAS modulates systemic RAS and how it is enhanced in obesity, which will explain the simultaneous development of metabolic syndrome alterations. Finally, dietary interventions that prevent obesity and adipocyte dysfunction will maintain normal RAS concentrations and effects, thus preventing metabolic diseases that are associated with RAS enhancement.
Collapse
|
14
|
Catalioto RM, Valenti C, Liverani L, Giuliani S, Maggi CA. Characterization of a novel proinflammatory effect mediated by BK and the kinin B₂ receptor in human preadipocytes. Biochem Pharmacol 2013; 86:508-20. [PMID: 23796753 DOI: 10.1016/j.bcp.2013.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/05/2013] [Accepted: 06/06/2013] [Indexed: 12/16/2022]
Abstract
Obesity and adipose tissue contribute to local and systemic inflammation. However the role of the inflammatory mediator bradykinin (BK) in this context is not known. We therefore evaluated the effect of BK on adipokines secretion in human preadipocytes during the course of differentiation and characterized the receptors involved. Results obtained from antibody array and ELISA experiments showed that several adipokines are released by human preadipocytes under basal conditions while BK specifically stimulated the production of interleukin(IL)-6 and IL-8. The effect of BK diminished with the progression of differentiation, being almost inactive on adipocytes. In preadipocytes, BK also induced a rapid and transient [Ca²⁺](i) mobilization, a rapid and sustained increase in ERK1/2 activation and enhanced forskolin-stimulated cAMP accumulation. BK was without effect on cell proliferation and viability as assessed by bromodeoxyuridine incorporation, WST-1 conversion, or lactate dehydrogenase leakage and was without effect on adipogenesis as measured by triglyceride accumulation, GPDH activity and leptin release. The B₁ receptor agonist, Lys-[des-Arg⁹]-BK, displayed poor activity or was without effect while overall BK effects were prevented by the selective B₂ receptor antagonist, fasitibant chloride, but not by the B₁ selective antagonist, Lys-[Leu⁸][des-Arg⁹]-BK. Immunoblot analysis and immunofluorescence studies showed that the kinin B₂ receptor was essentially expressed at the beginning of the differentiation program. In conclusion, human preadipocytes expressed kinin B₂ receptors linked to multiple signaling pathways, IL-6 and IL-8 production, and BK proinflammatory response in adipose tissue could be prevented by fasitibant chloride.
Collapse
Affiliation(s)
- Rose-Marie Catalioto
- Pharmacology Department, Menarini Ricerche SpA, Via Rismondo 12A, 50131 Florence, Italy.
| | | | | | | | | |
Collapse
|
15
|
Barros CC, Haro A, Russo FJ, Schadock I, Almeida SS, Reis FC, Moraes MR, Haidar A, Hirata AE, Mori M, Bacurau RFP, Würtele M, Bader M, Pesquero JB, Araujo RC. Bradykinin inhibits hepatic gluconeogenesis in obese mice. J Transl Med 2012; 92:1419-27. [PMID: 22868909 DOI: 10.1038/labinvest.2012.105] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The kallikrein-kinin system (KKS) has been previously linked to glucose homeostasis. In isolated muscle or fat cells, acute bradykinin (BK) stimulation was shown to improve insulin action and increase glucose uptake by promoting glucose transporter 4 translocation to plasma membrane. However, the role for BK in the pathophysiology of obesity and type 2 diabetes remains largely unknown. To address this, we generated genetically obese mice (ob/ob) lacking the BK B2 receptor (obB2KO). Despite similar body weight or fat accumulation, obB2KO mice showed increased fasting glycemia (162.3 ± 28.2 mg/dl vs 85.3 ± 13.3 mg/dl), hyperinsulinemia (7.71 ± 1.75 ng/ml vs 4.09 ± 0.51 ng/ml) and impaired glucose tolerance when compared with ob/ob control mice (obWT), indicating insulin resistance and impaired glucose homeostasis. This was corroborated by increased glucose production in response to a pyruvate challenge. Increased gluconeogenesis was accompanied by increased hepatic mRNA expression of forkhead box protein O1 (FoxO1, four-fold), peroxisome proliferator-activated receptor gamma co-activator 1-alpha (seven-fold), phosphoenolpyruvate carboxykinase (PEPCK, three-fold) and glucose-6-phosphatase (eight-fold). FoxO1 nuclear exclusion was also impaired, as the obB2KO mice showed increased levels of this transcription factor in the nucleus fraction of liver homogenates during random feeding. Intraportal injection of BK in lean mice was able to decrease the hepatic mRNA expression of FoxO1 and PEPCK. In conclusion, BK modulates glucose homeostasis by affecting hepatic glucose production in obWT. These results point to a protective role of the KKS in the pathophysiology of type 2 diabetes mellitus.
Collapse
|
16
|
Mori MA, Sales VM, Motta FL, Fonseca RG, Alenina N, Guadagnini D, Schadock I, Silva ED, Torres HAM, dos Santos EL, Castro CH, D’Almeida V, Andreotti S, Campaña AB, Sertié RAL, Saad MJA, Lima FB, Bader M, Pesquero JB. Kinin B1 receptor in adipocytes regulates glucose tolerance and predisposition to obesity. PLoS One 2012; 7:e44782. [PMID: 23024762 PMCID: PMC3443087 DOI: 10.1371/journal.pone.0044782] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 08/13/2012] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Kinins participate in the pathophysiology of obesity and type 2 diabetes by mechanisms which are not fully understood. Kinin B(1) receptor knockout mice (B(1) (-/-)) are leaner and exhibit improved insulin sensitivity. METHODOLOGY/PRINCIPAL FINDINGS Here we show that kinin B(1) receptors in adipocytes play a role in controlling whole body insulin action and glucose homeostasis. Adipocytes isolated from mouse white adipose tissue (WAT) constitutively express kinin B(1) receptors. In these cells, treatment with the B(1) receptor agonist des-Arg(9)-bradykinin improved insulin signaling, GLUT4 translocation, and glucose uptake. Adipocytes from B(1) (-/-) mice showed reduced GLUT4 expression and impaired glucose uptake at both basal and insulin-stimulated states. To investigate the consequences of these phenomena to whole body metabolism, we generated mice where the expression of the kinin B(1) receptor was limited to cells of the adipose tissue (aP2-B(1)/B(1) (-/-)). Similarly to B(1) (-/-) mice, aP2-B(1)/B(1) (-/-) mice were leaner than wild type controls. However, exclusive expression of the kinin B(1) receptor in adipose tissue completely rescued the improved systemic insulin sensitivity phenotype of B(1) (-/-) mice. Adipose tissue gene expression analysis also revealed that genes involved in insulin signaling were significantly affected by the presence of the kinin B(1) receptor in adipose tissue. In agreement, GLUT4 expression and glucose uptake were increased in fat tissue of aP2-B(1)/B(1) (-/-) when compared to B(1) (-/-) mice. When subjected to high fat diet, aP2-B(1)/B(1) (-/-) mice gained more weight than B(1) (-/-) littermates, becoming as obese as the wild types. CONCLUSIONS/SIGNIFICANCE Thus, kinin B(1) receptor participates in the modulation of insulin action in adipocytes, contributing to systemic insulin sensitivity and predisposition to obesity.
Collapse
Affiliation(s)
- Marcelo A. Mori
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | | | - Fabiana Louise Motta
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Raphael Gomes Fonseca
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Natalia Alenina
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Dioze Guadagnini
- Department of Internal Medicine, State University of Campinas, Campinas, São Paulo, Brazil
| | - Ines Schadock
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Elton Dias Silva
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | - Hugo A. M. Torres
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
| | | | | | - Vânia D’Almeida
- Department of Biosciences, Federal University of São Paulo, São Paulo, Brazil
| | - Sandra Andreotti
- Department of Physiology, University of São Paulo, São Paulo, São Paulo, Brazil
| | | | | | - Mario J. A. Saad
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Fabio Bessa Lima
- Department of Physiology, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - João Bosco Pesquero
- Department of Biophysics, Federal University of São Paulo, São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
17
|
Altered glucose homeostasis and hepatic function in obese mice deficient for both kinin receptor genes. PLoS One 2012; 7:e40573. [PMID: 22829877 PMCID: PMC3400662 DOI: 10.1371/journal.pone.0040573] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 06/09/2012] [Indexed: 12/22/2022] Open
Abstract
The Kallikrein-Kinin System (KKS) has been implicated in several aspects of metabolism, including the regulation of glucose homeostasis and adiposity. Kinins and des-Arg-kinins are the major effectors of this system and promote their effects by binding to two different receptors, the kinin B2 and B1 receptors, respectively. To understand the influence of the KKS on the pathophysiology of obesity and type 2 diabetes (T2DM), we generated an animal model deficient for both kinin receptor genes and leptin (obB1B2KO). Six-month-old obB1B2KO mice showed increased blood glucose levels. Isolated islets of the transgenic animals were more responsive to glucose stimulation releasing greater amounts of insulin, mainly in 3-month-old mice, which was corroborated by elevated serum C-peptide concentrations. Furthermore, they presented hepatomegaly, pronounced steatosis, and increased levels of circulating transaminases. This mouse also demonstrated exacerbated gluconeogenesis during the pyruvate challenge test. The hepatic abnormalities were accompanied by changes in the gene expression of factors linked to glucose and lipid metabolisms in the liver. Thus, we conclude that kinin receptors are important for modulation of insulin secretion and for the preservation of normal glucose levels and hepatic functions in obese mice, suggesting a protective role of the KKS regarding complications associated with obesity and T2DM.
Collapse
|
18
|
Schweitzer GG, Cartee GD. Postexercise skeletal muscle glucose transport is normal in kininogen-deficient rats. Med Sci Sports Exerc 2011; 43:1148-53. [PMID: 21200341 DOI: 10.1249/mss.0b013e31820a7f65] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
UNLABELLED A single exercise bout stimulates skeletal muscle glucose transport (GT) in the absence or presence of insulin. It has been suggested that the kallikrein-kinin system may contribute to exercise effects on both insulin-independent and insulin-dependent GT. Plasma kininogen, a key kallikrein-kinin system component, is a protein substrate for the enzyme kallikrein and the source of the peptide bradykinin. PURPOSE This study aimed to determine whether the postexercise (PEX) increase in insulin-dependent or insulin-independent GT is reduced in rats deficient in plasma kininogen versus normal rats. METHODS Male Brown Norway (BN) and Brown Norway Katholiek (BNK; plasma kininogen-deficient) rats were studied. BN and BNK rats were assigned to exercise (4×30-min swim) or sedentary (SED) groups. Rats were anesthetized immediately (0hPEX) or 3 h (3hPEX) after exercise. For 0hPEX and 0hSED rats, one epitrochlearis muscle per rat was used for AMPK phosphorylation and muscle glycogen analyses. The contralateral muscle was incubated with [H]-3-O-methylglucose (3-MG) for GT assay. For 3hPEX and 3hSED rats, one muscle from each rat was incubated without insulin, and the contralateral muscle was incubated with 60 μU·mL insulin, and both muscles were incubated with 3-MG for GT measurement. RESULTS For 0hPEX versus 0hSED, both BN and BNK rats had greater insulin-independent GT and AMPK phosphorylation with reduced glycogen after exercise. No genotype effects were found 0hPEX. There was a significant main effect of exercise (3hPEX>3hSED) and no interaction between exercise and genotype for basal or insulin-stimulated GT. CONCLUSIONS Plasma kininogen deficiency did not alter insulin-independent GT, AMPK phosphorylation, or glycogen depletion 0hPEX or insulin-dependent GT 3hPEX, suggesting that normal plasma kininogen is not essential for these important exercise effects.
Collapse
Affiliation(s)
- George G Schweitzer
- Muscle Biology Laboratory, School of Kinesiology, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
19
|
Schweitzer GG, Castorena CM, Hamada T, Funai K, Arias EB, Cartee GD. The B2 receptor of bradykinin is not essential for the post-exercise increase in glucose uptake by insulin-stimulated mouse skeletal muscle. Physiol Res 2011; 60:511-9. [PMID: 21401298 DOI: 10.33549/physiolres.932085] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Bradykinin can enhance skeletal muscle glucose uptake (GU), and exercise increases both bradykinin production and muscle insulin sensitivity, but bradykinin's relationship with post-exercise insulin action is uncertain. Our primary aim was to determine if the B2 receptor of bradykinin (B2R) is essential for the post-exercise increase in GU by insulin-stimulated mouse soleus muscles. Wildtype (WT) and B2R knockout (B2RKO) mice were sedentary or performed 60 minutes of treadmill exercise. Isolated soleus muscles were incubated with [³H]-2-deoxyglucose +/-insulin (60 or 100 microU/ml). GU tended to be greater for WT vs. B2RKO soleus with 60 microU/ml insulin (P=0.166) and was significantly greater for muscles with 100 microU/ml insulin (P<0.05). Both genotypes had significant exercise-induced reductions (P<0.05) in glycemia and insulinemia, and the decrements for glucose (approximately 14 %) and insulin (approximately 55 %) were similar between genotypes. GU tended to be greater for exercised vs. sedentary soleus with 60 microU/ml insulin (P=0.063) and was significantly greater for muscles with 100 microU/ml insulin (P<0.05). There were no significant interactions between genotype and exercise for blood glucose, plasma insulin or GU. These results indicate that the B2R is not essential for the exercise-induced decrements in blood glucose or plasma insulin or for the post-exercise increase in GU by insulin-stimulated mouse soleus muscle.
Collapse
Affiliation(s)
- G G Schweitzer
- Muscle Biology Laboratory, University of Michigan, School of Kinesiology, Ann Arbor, MI, USA
| | | | | | | | | | | |
Collapse
|
20
|
|
21
|
de Picoli Souza K, Batista EC, Silva ED, Reis FC, Silva SMA, Araujo RC, Luz J, Santos EL, Pesquero JB. Effect of kinin B2 receptor ablation on skeletal muscle development and myostatin gene expression. Neuropeptides 2010; 44:209-14. [PMID: 20045188 DOI: 10.1016/j.npep.2009.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2009] [Revised: 11/30/2009] [Accepted: 12/01/2009] [Indexed: 11/28/2022]
Abstract
Bradykinin (BK) is an active peptide that binds to the kinin B(2) receptor and induces biological events during the development and adult life. In this study we aimed to investigate the effect of kinin B(2) receptor ablation in the postnatal skeletal muscle development and body composition in adult life. For studies of skeletal muscle development, control (C57Bl6 - WT) and B(2) receptor knockout mice (B(2)(-/-)) were sacrificed at 15, 30 and 90days after birth, the gastrocnemius skeletal muscle was weighed and myostatin gene expression evaluated by real time PCR. For energy balance determination, data from control and B(2)(-/-) at 90 and 120days were collected by calorimetric method. Body composition at 120days was determined by chloroform-methanol (total body fat) and Lowry-modified method (total body protein). The results show that B(2)(-/-) have significantly increased total body weight at 15, 30 and 90days of life, when compared to WT. The weight of the gastrocnemius skeletal muscle was also significantly increased at 30 and 90days of life. Body composition analyses revealed that B(2)(-/-) mice exhibit more total corporal protein and less total corporal fat. Energy balance revealed that B(2)(-/-) have increased metabolizable energy intake and energy expenditure when compared to control mice, resulting in a lower energy gain. Interestingly, myostatin mRNA expression was significantly decreased in 15 and 30days old B(2)(-/-) mice and after icatibant treatment of WT adult mice for 5days. In conclusion, together our results show that kinin B(2) receptor deletion increases lean mass, reduces fat mass and improves metabolism efficiency in mice. The mechanism involved in this phenotype could be related to the reduction of myostatin gene expression during postnatal life.
Collapse
Affiliation(s)
- K de Picoli Souza
- School of Environmental and Biological Science, Federal University of Grande Dourados, Rodovia Dourados - Itahum, Km 12, 79804-970 Dourados, MS, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Respondek F, Swanson KS, Belsito KR, Vester BM, Wagner A, Istasse L, Diez M. Short-chain fructooligosaccharides influence insulin sensitivity and gene expression of fat tissue in obese dogs. J Nutr 2008; 138:1712-8. [PMID: 18716174 DOI: 10.1093/jn/138.9.1712] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Dietary fibers may modulate insulin resistance and glucose homeostasis in dogs. Their efficacy is, however, dependent on their origin, physical properties, and fermentability in the large bowel. Eight healthy Beagle dogs were fed a commercial diet at twice their maintenance requirements until they became obese. They were then maintained in the obese state and used in a cross-over design study to evaluate the effects of short-chain fructooligosaccharide (scFOS) supplementation (1% wt:wt dry matter in the diet). The euglycemic hyperinsulinemic clamp technique was performed before and after fattening and at the end of each 6-wk cross-over period. Fat tissue biopsies were taken in food-deprived and postprandial phases to measure mRNA abundance of genes involved with fatty acid, glucose metabolism, or inflammation. Insulin resistance appeared progressively with fattening and the rate of glucose infusion during euglycemic clamp was lower (P < 0.05) at the end of the fattening period (7.39 mg.kg(-1).min(-1)) than at baseline (21.21 mg.kg(-1).min(-1)). In stable obese dogs, scFOS increased (P < 0.05) the rate of glucose infusion compared with control (7.77 vs. 4.72 mg.kg(-1).min(-1)). Plasma insulin and triglyceride concentrations were greater in obese than in lean dogs but were not altered by scFOS. Whereas mRNA was not affected in food-deprived dogs, scFOS increased uncoupling protein 2 (P = 0.05) and tended to increase carnitine palmitoyl transferase 1 adipose mRNA levels during the postprandial period (P = 0.09). Adding 1% scFOS to the diet of obese dogs decreases insulin resistance and appears to modulate the transcription of genes involved in fatty acid or glucose metabolism.
Collapse
|
23
|
Fonseca-Alaniz MH, Takada J, Andreotti S, de Campos TBF, Campaña AB, Borges-Silva CN, Lima FB. High sodium intake enhances insulin-stimulated glucose uptake in rat epididymal adipose tissue. Obesity (Silver Spring) 2008; 16:1186-92. [PMID: 18369340 DOI: 10.1038/oby.2008.69] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE This study investigated the effect of different sodium content diets on rat adipose tissue carbohydrate metabolism and insulin sensitivity. METHODS AND PROCEDURES Male Wistar rats were fed on normal- (0.5% Na(+); NS), high- (3.12% Na(+); HS),or low-sodium (0.06% Na(+); LS) diets for 3, 6, and 9 weeks after weaning. Blood pressure (BP) was measured using a computerized tail-cuff system. An intravenous insulin tolerance test (ivITT) was performed in fasted animals. At the end of each period, rats were killed and blood samples were collected for glucose and insulin determinations. The white adipose tissue (WAT) from abdominal and inguinal subcutaneous (SC) and periepididymal (PE) depots were weighed and processed for adipocyte isolation and measurement of in vitro rates of insulin-stimulated 2-deoxy-D-[(3)H]-glucose uptake (2DGU) and conversion of -[U-(14)C]-glucose into (14)CO(2). RESULTS After 6 weeks, HS diet significantly increased the BP, SC and PE WAT masses, PE adipocyte size, and plasma insulin concentration. The sodium dietary content did not influence the whole-body insulin sensitivity. A higher half-maximal effective insulin concentration (EC(50)) from the dose-response curve of 2DGU and an increase in the insulin-stimulated glucose oxidation rate were observed in the isolated PE adipocytes from HS rats. DISCUSSION The chronic salt overload enhanced the adipocyte insulin sensitivity for glucose uptake and the insulin-induced glucose metabolization, contributing to promote adipocyte hypertrophy and increase the mass of several adipose depots, particularly the PE fat pad.
Collapse
MESH Headings
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Adipocytes, White/pathology
- Adipose Tissue/pathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Animals
- Biological Transport/drug effects
- Blood Glucose/metabolism
- Blood Pressure/drug effects
- Body Weight/physiology
- Cells, Cultured
- Dose-Response Relationship, Drug
- Epididymis/drug effects
- Epididymis/metabolism
- Epididymis/pathology
- Glucose/metabolism
- Glucose/pharmacokinetics
- Heart Rate/drug effects
- Hypertrophy
- Insulin/blood
- Insulin/pharmacology
- Male
- Rats
- Rats, Wistar
- Sodium, Dietary/pharmacology
Collapse
Affiliation(s)
- Miriam H Fonseca-Alaniz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
24
|
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
|
25
|
Dimitropoulou C, Chatterjee A, McCloud L, Yetik-Anacak G, Catravas JD. Angiotensin, bradykinin and the endothelium. Handb Exp Pharmacol 2007:255-94. [PMID: 16999222 DOI: 10.1007/3-540-32967-6_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Angiotensins and kinins are endogenous peptides with diverse biological actions; as such, they represent current and future targets of therapeutic intervention. The field of angiotensin biology has changed significantly over the last 50 years. Our original understanding of the crucial role of angiotensin II in the regulation of vascular tone and electrolyte homeostasis has been expanded to include the discovery of new angiotensins, their important role in cardiovascular inflammation and the development of clinically useful synthesis inhibitors and receptor antagonists. While less applied progress has been achieved in the kinin field, there are continuous discoveries in bradykinin physiology and in the complexity of kinin interactions with other proteins. The present review focuses on mechanisms and interactions of angiotensins and kinins that deal specifically with vascular endothelium.
Collapse
Affiliation(s)
- C Dimitropoulou
- Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912-2500, USA
| | | | | | | | | |
Collapse
|
26
|
Yuan G, Deng J, Wang T, Zhao C, Xu X, Wang P, Voltz JW, Edin ML, Xiao X, Chao L, Chao J, Zhang XA, Zeldin DC, Wang DW. Tissue kallikrein reverses insulin resistance and attenuates nephropathy in diabetic rats by activation of phosphatidylinositol 3-kinase/protein kinase B and adenosine 5'-monophosphate-activated protein kinase signaling pathways. Endocrinology 2007; 148:2016-26. [PMID: 17272402 PMCID: PMC2084357 DOI: 10.1210/en.2006-0602] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We previously reported that iv delivery of the human tissue kallikrein (HK) gene reduced blood pressure and plasma insulin levels in fructose-induced hypertensive rats with insulin resistance. In the current study, we evaluated the potential of a recombinant adeno-associated viral vector expressing the HK cDNA (rAAV-HK) as a sole, long-term therapy to correct insulin resistance and prevent renal damage in streptozotocin-induced type-2 diabetic rats. Administration of streptozotocin in conjunction with a high-fat diet induced systemic hypertension, diabetes, and renal damage in rats. Delivery of rAAV-HK resulted in a long-term reduction in blood pressure, and fasting plasma insulin was significantly lower in the rAAV-HK group than in the control group. The expression of phosphatidylinositol 3-kinase p110 catalytic subunit and the levels of phosphorylation at residue Thr-308 of Akt, insulin receptor B, and AMP-activated protein kinases were significantly decreased in organs from diabetic animals. These changes were significantly attenuated after rAAV-mediated HK gene therapy. Moreover, rAAV-HK significantly decreased urinary microalbumin excretion, improved creatinine clearance, and increased urinary osmolarity. HK gene therapy also attenuated diabetic renal damage as assessed by histology. Together, these findings demonstrate that rAAV-HK delivery can efficiently attenuate hypertension, insulin resistance, and diabetic nephropathy in streptozotocin-induced diabetic rats.
Collapse
Affiliation(s)
- Gang Yuan
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
| | - Juanjuan Deng
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
| | - Tao Wang
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
| | - Chunxia Zhao
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
| | - Xizheng Xu
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
| | - Peihua Wang
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
| | - James W. Voltz
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC USA
| | - Matthew L. Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC USA
| | - Xiao Xiao
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
- Departments of Molecular Genetics and Biochemistry & Gene Therapy Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lee Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Julie Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Xin A. Zhang
- Vascular Biology Center and Department of Medicine and Department of Molecular Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Darryl C. Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC USA
| | - Dao Wen Wang
- Department of Internal Medicine and Gene Therapy Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, People's Rep. of China
| |
Collapse
|
27
|
Abe KC, Mori MADS, Pesquero JB. Leptin deficiency leads to the regulation of kinin receptors expression in mice. ACTA ACUST UNITED AC 2006; 138:56-8. [PMID: 17184856 DOI: 10.1016/j.regpep.2006.11.018] [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] [Received: 03/30/2006] [Revised: 10/16/2006] [Accepted: 11/10/2006] [Indexed: 11/18/2022]
Abstract
Kinins are vasoactive and pro-inflammatory peptides generated by the cleavage of the kininogen by kallikreins. Two kinin receptors have been described and denominated B1 and B2. Obesity frequently accompanies other pathologies, such as diabetes and hypertention. The clustering of these pathologies is usually known as "metabolic syndrome". Mice lacking leptin gene (ob/ob) are severely obese and hyperphagic. Using quantitative RT-PCR analysis of B1 and B2 mRNAs expression, we described for the first time a correlation between the kallikrein-kinin system (KKS) and severe obesity in mice. The ob/ob mice presented lower expression of B2 mRNA in the white adipose tissue (WAT) and hypothalamus, both primary sites for neuroendocrine regulation of the energetic metabolism. B1 mRNA, however, is overexpressed in these tissues of ob/ob mice. An upregulation of the B1 mRNA has also been seen in liver, abdominal aorta and stomach fundus. However, different from the lean mice, the expression of the B1 mRNA in brown adipose tissue (BAT) and heart is completely abolished. Our data show that kinin receptors are differently modulated in distinct tissues in obesity. These findings suggest a connection between the KKS and obesity, and suggest that kinin receptors could be involved in the ethiopathogenesis of the metabolic syndrome.
Collapse
Affiliation(s)
- Karina Camasmie Abe
- Department of Biophysics, Federal University of São Paulo, 04023-062, São Paulo, Brazil
| | | | | |
Collapse
|
28
|
Beard KM, Lu H, Ho K, Fantus IG. Bradykinin augments insulin-stimulated glucose transport in rat adipocytes via endothelial nitric oxide synthase-mediated inhibition of Jun NH2-terminal kinase. Diabetes 2006; 55:2678-87. [PMID: 17003331 DOI: 10.2337/db05-1538] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
An increase in bradykinin has been suggested to contribute to the enhanced insulin sensitivity observed in the presence of ACE inhibitors. To investigate a potential direct, nonvascular effect on an insulin target tissue, the effect of bradykinin on glucose uptake and insulin signaling was studied in primary rat adipocytes. Whereas basal glucose uptake was not altered, bradykinin augmented insulin-stimulated glucose uptake twofold, which was blocked by HOE-140, a bradykinin B2 receptor antagonist. The bradykinin effect on glucose uptake was nitric oxide (NO) dependent, mimicked by NO donors and absent in adipocytes from endothelial NO synthase-/- mice. Investigation of insulin signaling revealed that bradykinin enhanced insulin receptor substrate-1 (IRS-1) Tyr phosphorylation, Akt/protein kinase B phosphorylation, and GLUT4 translocation. In contrast, insulin-stimulated extracellular signal-regulated kinase1/2 and Jun NH2-terminal kinase (JNK) activation were decreased in the presence of bradykinin, accompanied by decreased IRS-1 Ser307 phosphorylation. Furthermore, bradykinin did not enhance insulin action in the presence of the JNK inhibitor, SP-600125, or in adipocytes from JNK1-/- mice. These data indicate that bradykinin enhances insulin sensitivity in adipocytes via an NO-dependent pathway that acts by modulating the feedback inhibition of insulin signaling at the level of IRS-1.
Collapse
Affiliation(s)
- Kristin M Beard
- Department of Medicine and Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada M5G 1X6
| | | | | | | |
Collapse
|
29
|
Abstract
EH (essential hypertension) is a major public health problem in many countries due to its high prevalence and its association with coronary heart disease, stroke, renal disease, peripheral vascular disease and other disorders. Epidemiological studies have demonstrated that EH is heritable. Owing to the fact that blood pressure is controlled by cardiac output and total peripheral resistance, many molecular pathways are believed to be involved in the disease. In this review, recent genetic studies investigating the molecular basis of EH, including different molecular pathways, will be highlighted.
Collapse
Affiliation(s)
- Maolian Gong
- Max-Delbrück-Center for Molecular Medicine (MDC), Robert-Rössle-Strasse 10, 13092 Berlin, Germany
| | | |
Collapse
|
30
|
Spillmann F, Van Linthout S, Schultheiss HP, Tschöpe C. Cardioprotective mechanisms of the kallikrein-kinin system in diabetic cardiopathy. Curr Opin Nephrol Hypertens 2006; 15:22-9. [PMID: 16340662 DOI: 10.1097/01.mnh.0000199009.56799.2b] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE OF REVIEW Multiple pathogenic mechanisms contribute to the development of diabetic cardiopathy, including intramyocardial inflammation, cardiac fibrosis, abnormal intracellular Ca handling, microangiopathy and endothelial dysfunction. Moreover, the cardiac kallikrein-kinin system is thought to be altered under diabetic conditions and an improvement of this peptide system, e.g. by gene therapeutic approaches, has also been associated with an amelioration of the diabetic heart. In this review, we will discuss the hypothesis that the stimulation of the kallikrein-kinin system could be a promising target for the treatment of diabetic cardiopathy. RECENT FINDINGS The kallikrein-kinin system has cardioprotective properties, which may be particularly important under diabetic conditions. For example, its potential for endothelium-dependent vasodilation, and for improvement of glucose transport and utilization, make bradykinin an important mediator for reducing the consequences of diabetes-related oxidative stress on both the myocardium and vessels. SUMMARY The different synergistic cardioprotective effects of the kallikrein-kinin system in the diabetic heart suggest that the stimulation of the kallikrein-kinin system might open new avenues for the treatment of diabetic cardiopathy.
Collapse
Affiliation(s)
- Frank Spillmann
- Department of Cardiology and Pneumology, Charité-University Medicine of Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany
| | | | | | | |
Collapse
|
31
|
Araújo RC, Mori MA, Merino VF, Bascands JL, Schanstra JP, Zollner RL, Villela CA, Nakaie CR, Paiva ACM, Pesquero JL, Bader M, Pesquero JB. Role of the kinin B1 receptor in insulin homeostasis and pancreatic islet function. Biol Chem 2006; 387:431-6. [PMID: 16606341 DOI: 10.1515/bc.2006.057] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Kinins are potent vasoactive peptides generated in blood and tissues by the kallikrein serine proteases. Two distinct kinin receptors have been described, one constitutive (subtype B2) and one inducible (subtype B1), and many physiological functions have been attributed to these receptors, including glucose homeostasis and control of vascular permeability. In this study we show that mice lacking the kinin B1 receptor (B1
-/- mice) have lower fasting plasma glucose concentrations but exhibit higher glycemia after feeding when compared to wild-type mice. B1
-/- mice also present pancreas abnormalities, characterized by fewer pancreatic islets and lower insulin content, which leads to hypoinsulinemia and reduced insulin release after a glucose load. Nevertheless, an insulin tolerance test indicated higher sensitivity in B1
-/- mice. In line with this phenotype, pancreatic vascular permeability was shown to be reduced in B1 receptor-ablated mice. The B1 agonist desArg9bradykinin injected intravenously can induce the release of insulin into serum, and this effect was not observed in the B1
-/- mice or in isolated islets. Our data demonstrate the importance of the kinin B1 receptor in the control of pancreatic vascular homeostasis and insulin release, highlighting a new role for this receptor in the pathogenesis of diabetes and related diseases.
Collapse
|
32
|
Carter CS, Onder G, Kritchevsky SB, Pahor M. Angiotensin-converting enzyme inhibition intervention in elderly persons: effects on body composition and physical performance. J Gerontol A Biol Sci Med Sci 2006; 60:1437-46. [PMID: 16339331 DOI: 10.1093/gerona/60.11.1437] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The disablement process is often accompanied by sarcopenia or muscle loss, which is associated with virtually all identified disability risk factors. Clinically, the association between body composition and physical performance has been documented by several studies. However, loss of strength is greater than loss of muscle mass with age implying that the quality of remaining muscle may be reduced. Although there are limited data explaining potential physiological mechanisms that contribute to muscle quality, sarcopenia is frequently associated with fat accumulation, and the percentage of body fat increases with age even if weight does not. However, the relationship between fat and muscle function may not be linear, suggesting that there may be an optimal ratio of lean to fat mass for physical function. There are no definitive pharmacological interventions proven to prevent decline in physical function either by modulating body composition or by other means. One exception may be angiotensin-converting enzyme inhibitors (ACEIs). ACE is an important component of the renin-angiotensin system, the central hormonal regulator of blood pressure. Recent evidence suggests that ACEIs may improve physical function by means of direct effects on body composition in older persons, rather than through its blood-pressure-lowering effects. Clinical and genetic studies in humans and experimental evidence in animals suggest that modulation of the renin-angiotensin system is associated with metabolic and biochemical changes in skeletal muscle and fat, changes that are associated with declining physical function. ACEIs may modulate this process through a variety of molecular mechanisms including their influence on oxidative stress and on metabolic and inflammation pathways. This review describes potential biological mechanisms of ACE inhibition and its contribution to declining physical performance and changing body composition. Promising pharmacoepidemiological studies and experimental evidence in animals suggest that there are appropriate models in which to study this effect.
Collapse
Affiliation(s)
- Christy S Carter
- University of Florida, Department of Aging and Geriatric Research, 1329 SW 16th St. PO Box 100143, Gainesville FL, 32610-0143, USA.
| | | | | | | |
Collapse
|
33
|
Tomie Furuya D, Binsack R, Onishi ME, Monteiro Seraphim P, Fabres Machado U. Low ethanol consumption induces enhancement of insulin sensitivity in liver of normal rats. Life Sci 2005; 77:1813-24. [PMID: 15913658 DOI: 10.1016/j.lfs.2004.12.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Accepted: 12/21/2004] [Indexed: 01/01/2023]
Abstract
Moderate amounts of alcohol intake have been reported to have a protective effect on the cardiovascular system and this may involve enhanced insulin sensitivity. We established an animal model of increased insulin sensitivity by low ethanol consumption and here we investigated metabolic parameters and molecular mechanisms potentially involved in this phenomenon. For that, Wistar rats have received drinking water either without (control) or with 3% ethanol for four weeks. The effect of ethanol intake on insulin sensitivity was analyzed by insulin resistance index (HOMA-IR), intravenous insulin tolerance test (IVITT) and lipid profile. The role of liver was investigated by the analysis of insulin signaling pathway, GLUT2 gene expression and tissue glycogen content. Rats consuming 3% ethanol showed lower values of HOMA-IR and plasma free fatty acids (FFA) levels and higher hepatic glycogen content and glucose disappearance constant during the IVITT. Neither the phosphorylation of insulin receptor (IR) and insulin receptor substrate-1 (IRS-1), nor its association with phosphatidylinositol-3-kinase (PI3-kinase), was affected by ethanol. However, ethanol consumption enhanced liver IRS-2 and protein kinase B (Akt) phosphorylation (3 times, P<0.05), which can be involved in the 2-fold increased (P<0.05) hepatic glycogen content. The GLUT2 protein content was unchanged. Our findings point out that liver plays a role in enhanced insulin sensitivity induced by low ethanol consumption.
Collapse
Affiliation(s)
- Daniela Tomie Furuya
- Dept. of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo. Av. Prof. Lineu Prestes 1524, 05508-900 Sao Paulo, Brazil
| | | | | | | | | |
Collapse
|
34
|
Couture R, Girolami JP. Putative roles of kinin receptors in the therapeutic effects of angiotensin 1-converting enzyme inhibitors in diabetes mellitus. Eur J Pharmacol 2005; 500:467-85. [PMID: 15464053 DOI: 10.1016/j.ejphar.2004.07.045] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2004] [Indexed: 02/07/2023]
Abstract
The role of endogenous kinins and their receptors in diabetes mellitus is being confirmed with the recent developments of molecular and genetic animal models. Compelling evidence suggests that the kinin B(2) receptor is organ-protective and partakes to the therapeutic effects of angiotensin 1-converting enzyme inhibitors (ACEI) and angiotensin AT(1) receptor antagonists. Benefits derive primarily from vasodilatory, antihypertensive, antiproliferative, antihypertrophic, antifibrotic, antithrombotic and antioxidant properties of kinin B(2) receptor activation. Mechanisms include the formation of nitric oxide and prostacyclin and the inhibition of NAD(P)H oxidase activity involving classical and novel signalling pathways. Kinin B(2) receptor also ameliorates insulin resistance by increasing glucose uptake and supply, and by inducing glucose transporter-4 translocation either directly or through phosphorylation of insulin receptor. The kinin B(1) receptor, which is induced by the cytokine network, growth factors and hyperglycaemia, mediates hyperalgesia, vascular hyperpermeability and leukocytes infiltration in diabetic animals. However, emerging data highlight reno- and cardio-protective effects mediated by kinin B(1) receptor under chronic ACEI therapy in diabetes mellitus. Thus, the Janus-faced of kinin receptors needs to be taken into account in future drug development. For instance, locally acting kinin B(1)/B(2) receptor agonists if used in a safe therapeutic window may represent a more rationale strategy in the prevention and management of diabetic complications. Because kinin B(2) receptor antagonists may further increase insulin resistance, the persisting dogma that restricts the development of kinin receptor analogues to antagonists (that is still relevant to abrogate pain and inflammation) needs to be revisited.
Collapse
Affiliation(s)
- Réjean Couture
- Département de Physiologie, Faculté de Médecine, Université de Montréal, C.P. 6128, Succursale centre-ville, Montréal, Québec, Canada H3C 3J7.
| | | |
Collapse
|
35
|
Montanari D, Yin H, Dobrzynski E, Agata J, Yoshida H, Chao J, Chao L. Kallikrein gene delivery improves serum glucose and lipid profiles and cardiac function in streptozotocin-induced diabetic rats. Diabetes 2005; 54:1573-80. [PMID: 15855348 DOI: 10.2337/diabetes.54.5.1573] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We investigated the role of the kallikrein-kinin system in cardiac function and glucose utilization in the streptozotocin (STZ)-induced diabetic rat model using a gene transfer approach. Adenovirus harboring the human tissue kallikrein gene was administered to rats by intravenous injection at 1 week after STZ treatment. Human kallikrein transgene expression was detected in the serum and urine of STZ-induced diabetic rats after gene transfer. Kallikrein gene delivery significantly reduced blood glucose levels and cardiac glycogen accumulation in STZ-induced diabetic rats. Kallikrein gene transfer also significantly attenuated elevated plasma triglyceride and cholesterol levels, food and water intake, and loss of body weight gain, epididymal fat pad, and gastrocnemius muscle weight in STZ-induced diabetic rats. However, these effects were blocked by icatibant, a kinin B2 receptor antagonist. Cardiac function was significantly improved after kallikrein gene transfer as evidenced by increased cardiac output and +/-delta P/delta t (maximum speed of contraction/relaxation), along with elevated cardiac sarco(endo)plasmic reticulum (Ca2+ + Mg2+)-ATPase (SERCA)-2a, phosphorylated phospholamban, NOx and cAMP levels, and GLUT4 translocation into plasma membranes of cardiac and skeletal muscle. Kallikrein gene delivery also increased Akt and glycogen synthase kinase (GSK)-3beta phosphorylation, resulting in decreased GSK-3beta activity in the heart. These results indicate that kallikrein through kinin formation protects against diabetic cardiomyopathy by improving cardiac function and promoting glucose utilization and lipid metabolism.
Collapse
Affiliation(s)
- David Montanari
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425-2211, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Cui J, Melista E, Chazaro I, Zhang Y, Zhou X, Manolis AJ, Baldwin CT, Destefano AL, Gavras H. Sequence variation of bradykinin receptors B1 and B2 and association with hypertension. J Hypertens 2005; 23:55-62. [PMID: 15643125 DOI: 10.1097/00004872-200501000-00013] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To identify variants in the complete genomic sequence of the two subtypes of bradykinin receptors: B1 (BDKRB1) and B2 (BDKRB2) and to examine the association of these variants with essential hypertension. DESIGN A case-control design comparing hypertensive and normotensive individuals was used. METHOD A 64.4 kb genomic region containing the BDKRB1 and BDKRB2 genes was sequenced in 30 African-American individuals. A total of 282 single-nucleotide polymorphisms (SNPs) were identified. A total of 21 SNPs were genotyped in our complete cohorts of hypertensive and normotensive African-Americans (n = 218), American-Caucasians (n = 220) and Greek-Caucasians (n = 194). Pair-wise correlation coefficients were computed to assess linkage disequilibrium (LD) patterns among the SNPs. Chi-squared tests and logistic regression were used to assess association between the SNPs and hypertension status. RESULT Pairwise LD demonstrated a general pattern of decline with increasing distance, which was consistent among the three groups with less LD in African-Americans. One SNP in the promoter region of BDKRB2 (rs1799722) was associated with hypertension (P = 0.044) in African-Americans. One SNP in BDKRB2 and three SNPs in BDKRB1 were associated with hypertension (P-values between 0.026 and 0.0004) in American-Caucasians. Haplotypes including those four SNPs and one SNP in B2, which results in an amino acid change, demonstrated a significant haplotype frequency difference between hypertensive and normotensive American-Caucasians (P = 0.025). CONCLUSION These results support the hypothesis that the African-American population is an older population compared with the other samples and the two bradykinin receptors may play a role in blood pressure regulation.
Collapse
Affiliation(s)
- Jing Cui
- Hypertension Section, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Carter CS, Cesari M, Ambrosius WT, Hu N, Diz D, Oden S, Sonntag WE, Pahor M. Angiotensin-converting enzyme inhibition, body composition, and physical performance in aged rats. J Gerontol A Biol Sci Med Sci 2004; 59:416-23. [PMID: 15123750 DOI: 10.1093/gerona/59.5.b416] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study was designed to test the effects of angiotensin-converting enzyme (ACE) inhibition on body composition and physical performance in aged rats. Male Brown Norway x F344 rats were randomized to receive daily injections of enalapril (40 mg/kg or 80 mg/kg) or saline from 24 to 30 months of age. Body composition was determined using dual-energy X-ray absorptiometry (DXA), and physical performance was assessed using the grip strength and inclined plane procedures. Performance measures were assessed at baseline and monthly thereafter. DXA was performed at baseline, 3 months, and 6 months of follow-up. Compared with the enalapril groups, the saline group experienced a greater 6-month decline in the physical performance measures. Lean body mass declined in both groups; however, the enalapril groups also experienced a significant loss of fat mass. These results suggest that ACE inhibition may prevent age-related declines in physical performance, which may be mediated by a reduction in body fat mass.
Collapse
Affiliation(s)
- Christy S Carter
- Sticht Center on Aging, Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Okamoto MM, Sumida DH, Carvalho CRO, Vargas AM, Heimann JC, Schaan BD, Machado UF. Changes in dietary sodium consumption modulate GLUT4 gene expression and early steps of insulin signaling. Am J Physiol Regul Integr Comp Physiol 2004; 286:R779-85. [PMID: 14670806 DOI: 10.1152/ajpregu.00396.2003] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies have shown that chronic salt overload increases insulin sensitivity, while chronic salt restriction decreases it. In the present study we investigated the influence of dietary sodium on 1) GLUT4 gene expression, by Northern and Western blotting analysis; 2) in vivo GLUT4 protein translocation, by measuring the GLUT4 protein in plasma membrane and microsome, before and after insulin injection; and 3) insulin signaling, by analyzing basal and insulin-stimulated tyrosine phosphorylation of insulin receptor (IR)-β, insulin receptor substrate (IRS)-1, and IRS-2. Wistar rats were fed normal-sodium (NS-0.5%), low-sodium (LS-0.06%), or high-sodium diets (HS-3.12%) for 9 wk and were killed under pentobarbital anesthesia. Compared with NS rats, HS rats increased ( P < 0.05) the GLUT4 protein in adipose tissue and skeletal muscle, whereas GLUT4 mRNA was increased only in adipose tissue. GLUT4 expression was unchanged in LS rats compared with NS rats. The GLUT4 translocation in HS rats was higher ( P < 0.05) both in basal and insulin-stimulated conditions. On the other hand, LS rats did not increase the GLUT4 translocation after insulin stimulus. Compared with NS rats, LS rats showed reduced ( P < 0.01) basal and insulin-stimulated tyrosine phosphorylation of IRS-1 in skeletal muscle and IRS-2 in liver, whereas HS rats showed enhanced basal tyrosine phosphorylation of IRS-1 in skeletal muscle ( P < 0.05) and of IRS-2 in liver. In summary, increased insulin sensitivity in HS rats is related to increased GLUT4 gene expression, enhanced insulin signaling, and GLUT4 translocation, whereas decreased insulin sensitivity of LS rats does not involve changes in GLUT4 gene expression but is related to impaired insulin signaling.
Collapse
Affiliation(s)
- Maristela Mitiko Okamoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 05508-900, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
39
|
Cahová M, Vavrinková H, Tutterova M, Meschisvilli E, Kazdova L. Captopril enhanced insulin-stimulated glycogen synthesis in skeletal muscle but not fatty acid synthesis in adipose tissue of hereditary hypertriglyceridemic rats. Metabolism 2003; 52:1406-12. [PMID: 14624398 DOI: 10.1016/s0026-0495(03)00319-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In addition to their hypotensive action, angiotensin-converting enzyme (ACE) inhibitors exert a beneficial effect on glucoregulation. In the present study, the effect of ACE inhibition by captopril on glucose utilization in peripheral tissues was investigated in non-obese rats with hereditary hypertriglyceridemia (HHTg) associated with hyperinsulinemia and insulin resistance. Normotriglyceridemic Wistar rats served as controls (C). Rats of both groups received a high-sucrose diet, and a half of each group also captopril in drinking water (10 mg/kg body weight [bw]) for 2 weeks. Captopril administration reduced fasting glycemia and postprandial triglyceridemia in HHTg rats, while the fasting levels of nonesterified fatty acids (NEFA), glycerol, and lactate were decreased in both groups. The sensitivity of skeletal muscle to insulin action evaluated as in vitro 14C-glucose incorporation into glycogen was significantly increased by captopril treatment both in HHTg (3.51 +/- 0.48 v 2.0 +/- 0.12 micromol glucose/g wet weight [ww]) and C (3.32 +/- 0.21 v 2.48 +/- 0.09 micromol glucose/g ww). In isolated adipose tissue, the insulin-stimulated 14C-glucose incorporation into neutral lipids was increased, after captopril administration, by 137% in C and by 35% only in HHTg. After captopril treatment, insulin-stimulated de novo fatty acid synthesis rose significantly in C while remaining low in HHTg. The increase in esterification was comparable in both groups. Separate experiments were designed to assess the possible involvement of bradykinin in mediating captopril action. Both C and HHTg rats fed a high-sucrose diet for 2 weeks were treated with captopril (50 mg/kg orally) for 1 hour; half of each group received the specific inhibitor of bradykinin receptor HOE-140 (100 microg/kg intraperitoneally [i.p.]) 1 hour before captopril administration. In C, captopril administration enhanced the insulin-stimulated in vitro glucose incorporation into lipids in adipose tissue by 255%, and into glycogen in the musculus soleus by 45%; this effect was eliminated by HOE-140. In HHTg, neither a single dose of captopril nor HOE-140 had any effect. We conclude that long-term captopril administration increased the insulin sensitivity of peripheral tissue in both C and HHTg rats, but with different efficacy. While the insulin-sensitizing action of captopril on skeletal muscle was comparable in HHTg and C rats, there were differences in the effect of captopril on adipose tissue. The difference became particularly manifest in de novo fatty acid synthesis.
Collapse
Affiliation(s)
- Monika Cahová
- Department of Metabolic Research, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
40
|
McCarty MF. ACE inhibition may decrease diabetes risk by boosting the impact of bradykinin on adipocytes. Med Hypotheses 2003; 60:779-83. [PMID: 12699703 DOI: 10.1016/s0306-9877(02)00234-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The findings of the recent HOPE trial strongly suggest that ACE inhibitor therapy may reduce risk for type 2 diabetes in patients who are non-diabetic at baseline. This finding is readily rationalized by previous evidence that bradykinin, acting via B2 receptors, can potentiate the insulin responsiveness of both adipocytes and muscle fibers; this effect may be mediated by a reduction in the activity of a tyrosine phosphatase that targets the insulin receptor. ACE inhibitors, in turn, increase the availability of bradykinin by suppressing its proteolytic degradation. In light of the fact that the development of insulin resistance in adipocytes is responsible for the excessive free fatty acid flux that gives rise to the diabetic syndrome, a favorable impact of ACE inhibition on adipocyte insulin responsiveness - complemented by a potentiation of the direct action of bradykinin on skeletal muscle - offers a satisfying explanation for the prevention of diabetes observed during ACE inhibitor therapy. Since the population at risk for diabetes is huge and increasing dramatically, the recent development of orally absorbable food-derived peptides with clinically significant ACE inhibitory activity - such as 'Katsuobushi oligopeptides' derived from bonito - may make it more logistically feasible to achieve this protection on a widescale basis, while simultaneously promoting blood pressure control and reducing risk for atherothrombotic disease.
Collapse
Affiliation(s)
- M F McCarty
- Pantox Laboratories, San Diego, California 92109, USA
| |
Collapse
|
41
|
|
42
|
Ogihara T, Asano T, Ando K, Sakoda H, Anai M, Shojima N, Ono H, Onishi Y, Fujishiro M, Abe M, Fukushima Y, Kikuchi M, Fujita T. High-salt diet enhances insulin signaling and induces insulin resistance in Dahl salt-sensitive rats. Hypertension 2002; 40:83-9. [PMID: 12105143 DOI: 10.1161/01.hyp.0000022880.45113.c9] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A high-salt diet, which is known to contribute to the pathogenesis of hypertension, is also reportedly associated with insulin resistance. We investigated the effects of a high-salt diet on insulin sensitivity and insulin signaling in salt-sensitive (Dahl-S) and salt resistant (Dahl-R) strains of the Dahl rat. Evaluation of hyperinsulinemic-euglycemic clamp studies and glucose uptake into the isolated soleus muscle revealed that salt loading (8% NaCl) for 4 weeks induced hypertension and significant insulin resistance in Dahl-S rats, whereas no significant effects were observed in Dahl-R rats. Despite the presence of insulin resistance, insulin-induced tyrosine phosphorylation of the insulin receptor and insulin receptor substrates, activation of phosphatidylinositol 3-kinase, and phosphorylation of Akt were all enhanced in Dahl-S rats fed a high-salt diet. The mechanism underlying this form of insulin resistance thus differs from that previously associated with obesity and dexamethasone and is likely due to the impairment of one or more metabolic steps situated downstream of phosphatidylinositol 3-kinase and Akt activation. Interestingly, supplementation of potassium (8% KCl) ameliorated the changes in insulin sensitivity in Dahl-S rats fed a high-salt diet; this was associated with a slight but significant decrease in blood pressure. Evidence presented suggest that there is an interdependent relationship between insulin sensitivity and salt sensitivity of blood pressure in Dahl-S rats, and it is suggested that supplementing the diet with potassium may exert a protective effect against both hypertension and insulin resistance in salt-sensitive individuals.
Collapse
Affiliation(s)
- Takehide Ogihara
- Department of Internal Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dumke CL, Kim J, Arias EB, Cartee GD. Role of kallikrein-kininogen system in insulin-stimulated glucose transport after muscle contractions. J Appl Physiol (1985) 2002; 92:657-64. [PMID: 11796678 DOI: 10.1152/japplphysiol.00854.2001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Serum proteins [molecular weight (MW) > 10,000] are essential for increased insulin-stimulated glucose transport after in vitro muscle contractions. We investigated the role of the kallikrein-kininogen system, including bradykinin, which is derived from kallikrein (MW > 10,000)-catalyzed degradation of serum protein kininogen (MW > 10,000), on this contraction effect. In vitro electrical stimulation of rat epitrochlearis muscles was performed in 1) rat serum +/- kallikrein inhibitors; 2) human plasma (normal or kallikrein-deficient); 3) rat serum +/- bradykinin receptor-2 inhibitors; or 4) serum-free buffer +/- bradykinin. 3-O-methylglucose transport (3-MGT) was measured 3.5 h later. Serum +/- kallikrein inhibitors tended (P = 0.08) to diminish postcontraction insulin-stimulated 3-MGT. Contractions in normal plasma enhanced insulin-stimulated 3-MGT vs. controls, but contractions in kallikrein-deficient plasma did not. Supplementing rat serum with bradykinin receptor antagonist HOE-140 during contraction did not alter insulin-stimulated 3-MGT. Muscles stimulated to contract in serum-free buffer plus bradykinin did not have enhanced insulin-stimulated 3-MGT. Bradykinin was insufficient for postcontraction-enhanced insulin sensitivity. However, results with kallikrein inhibitors and kallikrein-deficient plasma suggest kallikrein plays a role in this improved insulin action.
Collapse
Affiliation(s)
- C L Dumke
- Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin, Madison, Wisconsin 53706, USA.
| | | | | | | |
Collapse
|
44
|
Duka I, Shenouda S, Johns C, Kintsurashvili E, Gavras I, Gavras H. Role of the B(2) receptor of bradykinin in insulin sensitivity. Hypertension 2001; 38:1355-60. [PMID: 11751717 DOI: 10.1161/hy1201.096574] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The biological actions of bradykinin (BK) are attributed to its B(2) type receptor (B(2)R), whereas the B(1)R is constitutively absent, inducible by inflammation and toxins. Previous studies in B(2)R gene knockout mice showed that the B(1)R is overexpressed, is further upregulated by hypertensive maneuvers, and assumes some of the hemodynamic functions of the B(2)R. The current experiments were designed to further clarify the metabolic function of the B(2)R and to explore whether the upregulated B(1)R can also assume the metabolic function of the missing B(2)R. One group of B(2)R-/- mice (n=9) and one of B(2)R+/+ controls (n=8) were treated for 3 days with captopril (which produced a similar blood pressure-lowering response in both groups) and studied with the hyperinsulinemic euglycemic clamp. The knockout mice had fasting and steady-state blood glucose levels similar to those of the wild-type mice but a had tendency to higher fasting insulin levels (at 27.8+/-5.2 versus 18+/-2.9 mU/L, respectively). However, they had significantly higher steady-state insulin levels (749+/-127.2 versus 429.1+/-31.5 mU/L, P<0.05) and a significantly lower glucose uptake rate (31+/-2.4 versus 41+/-2.3 mg/kg per minute, P<0.05) and insulin sensitivity index (4.6+/-0.9 versus 10+/-0.7 P<0.001). Analysis of B(1)R and B(2)R gene expression by reverse transcription-polymerase chain reaction in cardiac muscle, skeletal muscle, and adipose tissues revealed significantly higher B(1)R mRNA level in the knockouts versus wild-type (P<0.05) at baseline and a further significant upregulation in mRNA by 1.8- to 3.2-fold (P<0.05) after insulin infusion. We conclude that absence of B(2)R confers a state of insulin resistance because it results in impaired insulin-dependent glucose transport; this is probably a direct B(2)R effect because, unlike the hemodynamic autacoid-mediated effects, it cannot be assumed by the upregulated B(1)R.
Collapse
Affiliation(s)
- I Duka
- Hypertension and Atherosclerosis Section of the Department of Medicine, Boston University School of Medicine, MA 02118, USA
| | | | | | | | | | | |
Collapse
|
45
|
Shiuchi T, Nakagami H, Iwai M, Takeda Y, Cui T, Chen R, Minokoshi Y, Horiuchi M. Involvement of bradykinin and nitric oxide in leptin-mediated glucose uptake in skeletal muscle. Endocrinology 2001; 142:608-12. [PMID: 11159831 DOI: 10.1210/endo.142.2.7964] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Regulation of glucose metabolism in peripheral tissues by leptin has been highlighted recently, although its mechanism is unclear. In this study, we postulated that bradykinin and nitric oxide (NO) are involved in the effect of leptin-mediated glucose uptake in peripheral tissues and examined these possibilities. Injection of leptin (200 pg/mouse) into the ventromedial hypothalamus-enhanced glucose uptake in skeletal muscle and brown adipose tissue, but not in white adipose tissue. Treatment with Hoe140 (0.1 mg/kg), bradykinin B2 receptor antagonist, or L-NAME (N:(G)-nitro-L-arginine methyl ester) (30 mg/kg), nitric oxide synthase inhibitor, did not influence the basal level of glucose uptake in skeletal muscle and the adipose tissue, whereas Hoe140 and L-NAME inhibited leptin-mediated glucose uptake in skeletal muscles, but had no effect in adipose tissue. However, Hoe140 and L-NAME did not inhibit insulin (1.0 U/kg)-mediated glucose uptake in all tissues examined. Taken together, these results suggest that leptin enhances bradykinin and/or the NO system, which contributes at least partially to the enhanced glucose uptake in skeletal muscles.
Collapse
Affiliation(s)
- T Shiuchi
- Department of Medical Biochemistry, Ehime University School of Medicine, Shigenobu, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Abstract
We investigated the effect of angiotensin-converting enzyme inhibitors on glucose uptake regulation as well as the effect of bradykinin (BK) on glucose uptake and its regulation by using inhibitors of phospholipase C, BK B2 receptor, protein kinase C, phosphatidylinositol 3-kinase, tyrosine kinase, and intracellular Ca(2+). We measured 2-deoxyglucose uptake by using L(6) skeletal muscle cells. In the presence of 1 nmol/L of insulin, 1 micromol/L of enalaprilat enhanced insulin-induced glucose uptake from 89.2+/-8. 1 to 138.0+/-13.6 pmol/h per mg protein. The stimulation of glucose uptake with enalaprilat was blocked to 92.7+/-7.8 pmol/h per mg protein by 10 micromol/L HOE 140 (a BK B2 receptor antagonist). In the presence of 1 nmol/L of insulin, exposure to 10 micromol/L BK stimulated glucose uptake from 89.2+/-8.1 to 171.6+/-10.1 pmol/h per mg protein. However, in the absence of insulin, BK could not enhance glucose uptake. One hundred nanomoles per liter of tyrphostin A-23 and genistein, which are tyrosine kinase inhibitors, significantly decreased the BK-induced glucose uptake from 142.0+/-8.4 to 87.6+/-6. 4 and 85.2+/-7.3 pmol/h per mg protein, respectively. BK-induced glucose uptake was inhibited significantly by 10 micromol/L U73122 (a phospholipase C antagonist) from 142.0+/-8.4 to 95.7+/-9.5 pmol/h per mg protein. One and 20 micromol/L of TMB-8 (an intracellular calcium antagonist) significantly decreased BK-induced glucose uptake from 142.0+/-8.4 to 108.0+/-9.6 and 100.8+/-11.4 pmol/h per mg protein. Angiotensin-converting enzyme inhibitors enhanced insulin-induced glucose uptake via the BK B2 receptor. BK-stimulated glucose uptake is related to phospholipase C, tyrosine kinase, and an increase in intracellular calcium.
Collapse
Affiliation(s)
- A Kudoh
- Department of Anesthesiology, University of Hirosaki School of Medicine, Aomori, Japan
| | | |
Collapse
|
48
|
Motoshima H, Araki E, Nishiyama T, Taguchi T, Kaneko K, Hirashima Y, Yoshizato K, Shirakami A, Sakai K, Kawashima J, Shirotani T, Kishikawa H, Shichiri M. Bradykinin enhances insulin receptor tyrosine kinase in 32D cells reconstituted with bradykinin and insulin signaling pathways. Diabetes Res Clin Pract 2000; 48:155-70. [PMID: 10802154 DOI: 10.1016/s0168-8227(00)00121-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have previously shown that bradykinin potentiated insulin-induced glucose uptake through GLUT4 translocation in canine adipocytes and skeletal muscles. The aim of this study was to determine the molecular mechanism of bradykinin enhancement of the insulin signal. For this purpose, 32D cells, which express a limited number of insulin receptors and lack endogenous bradykinin B2 receptor (BK2R) or insulin receptor substrate (IRS)-1 were transfected with BK2R cDNA and/or insulin receptor cDNA and/or IRS-1 cDNA, and analyzed. In 32D cells that expressed BK2R and insulin receptor (32D-BKR/IR), bradykinin alone had no effect on the phosphorylation of the insulin receptor, but it enhanced insulin-stimulated tyrosine phosphorylation of the insulin receptor. In 32D cells that expressed BK2R, insulin receptor and IRS-1 (32D-BKR/IR/IRS1), bradykinin also enhanced insulin-stimulated tyrosine phosphorylation of the insulin receptor and IRS-1. An increase in insulin-stimulated phosphorylation of IRS-1 by treatment with bradykinin in 32D-BKR/IR/IRS1 cell was associated with increased binding of 85 kD subunit of phosphatidylinositol 3 (PI 3)-kinase and increased IRS-1 associated PI 3-kinase activity. These effects of bradykinin were not observed in 32D cells which lack the expression of BK2R (32D-IR/IRS1) or insulin receptor (32D-BKR/IRS1). Furthermore, tyrosine phosphatase activity against insulin receptor beta-subunit in plasma membrane fraction of 32D-BKR/IR cells was significantly reduced by bradykinin, suggesting that the effect of bradykinin was in part mediated by inhibition of protein tyrosine phosphatase(s). Our results clearly demonstrated that bradykinin enhanced insulin-stimulated tyrosine kinase activity of the insulin receptor and downstream insulin signal cascade through the BK2R mediated signal pathway.
Collapse
Affiliation(s)
- H Motoshima
- Department of Metabolic Medicine, Kumamoto University School of Medicine, 1-1-1 Honjo, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Caldiz CI, de Cingolani GE. Insulin resistance in adipocytes from spontaneously hypertensive rats: effect of long-term treatment with enalapril and losartan. Metabolism 1999; 48:1041-6. [PMID: 10459571 DOI: 10.1016/s0026-0495(99)90203-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Insulin responsiveness was studied in isolated adipocytes from the normotensive Wistar Kyoto (WKY) rat and the spontaneously hypertensive rat (SHR). The effect of insulin (0.1 to 5 nmol/L) on glucose uptake (glucose transport and lipogenesis) was measured, and the maximal effect of insulin (Emax) and the dose of insulin required to elicit 50% of the maximal response (EC50) were calculated. A diminished Emax on lipogenesis without changes in the EC50 was detected in SHRs. The Emax was 0.49 +/- 0.09 (SHR) and 1.16 +/- 0.14 (WKY) micromol/10(5) cells (P < .05), and the EC50 was 0.13 +/- 0.03 and 0.11 +/- 0.02 nmol/L for WKY and SHR, respectively. Similar results were obtained when measuring insulin-stimulated glucose transport. A 30-day long-term treatment with enalapril (20 mg/kg/d) normalized insulin responsiveness in adipocytes from SHRs. The effect of enalapril was suppressed when SHRs were pretreated with enalapril and 150 microg/kg/d of the bradykinin (BK) B2-receptor blocker Hoe 140. Pretreatment with losartan (40 mg/kg/d) did not improve insulin action in the SHR. Since these results were obtained with isolated cells in which glucose availability was not a function of blood flow, and the effect of insulin in the SHR was improved by pretreatment with an angiotensin-converting enzyme (ACE) inhibitor but not with the AT1-receptor blocker, it appears that the insulin resistance linked to the hypertension is not related to changes in blood flow.
Collapse
Affiliation(s)
- C I Caldiz
- Center of Cardiovascular Research, School of Medicine, National University of La Plata, Argentina
| | | |
Collapse
|
50
|
Abstract
Peptide ligands carrying additional reporter groups are valuable research tools to facilitate biochemical and pharmacological studies of G protein-coupled receptors. B2 bradykinin receptors, widely distributed in mammalian tissues, regulate many physiological systems and are therapeutic targets. Acylation of the amino-terminus of bradykinin (BK) and a B2a-selective antagonist produced ligands derivatized with biotinamidocaproate or 7-Amino-4-methylcoumarin-3-acetate. These fluorescent and biotinylated peptides bound with high affinity to bovine and rodent B2 receptors. Analysis of second messenger production confirmed that fluorescent and biotinylated analogs of BK were B2 receptor agonists whereas derivatives of DArg0[Hyp3,DPhe7,Leu8]BK were BK receptor antagonists. The complimentary properties of these selective receptor probes will be useful in studying B2 receptor localization, expression and desensitization.
Collapse
Affiliation(s)
- J Howl
- Molecular Pharmacology Group, School of Health Sciences, University of Wolverhampton, UK.
| |
Collapse
|