1
|
Cousineau JP, Dawe AM, Alpaugh M. Investigating the Interplay between Cardiovascular and Neurodegenerative Disease. BIOLOGY 2024; 13:764. [PMID: 39452073 PMCID: PMC11505144 DOI: 10.3390/biology13100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024]
Abstract
Neurological diseases, including neurodegenerative diseases (NDDs), are the primary cause of disability worldwide and the second leading cause of death. The chronic nature of these conditions and the lack of disease-modifying therapies highlight the urgent need for developing effective therapies. To accomplish this, effective models of NDDs are required to increase our understanding of underlying pathophysiology and for evaluating treatment efficacy. Traditionally, models of NDDs have focused on the central nervous system (CNS). However, evidence points to a relationship between systemic factors and the development of NDDs. Cardiovascular disease and related risk factors have been shown to modify the cerebral vasculature and the risk of developing Alzheimer's disease. These findings, combined with reports of changes to vascular density and blood-brain barrier integrity in other NDDs, such as Huntington's disease and Parkinson's disease, suggest that cardiovascular health may be predictive of brain function. To evaluate this, we explore evidence for disruptions to the circulatory system in murine models of NDDs, evidence of disruptions to the CNS in cardiovascular disease models and summarize models combining cardiovascular disruption with models of NDDs. In this study, we aim to increase our understanding of cardiovascular disease and neurodegeneration interactions across multiple disease states and evaluate the utility of combining model systems.
Collapse
Affiliation(s)
| | | | - Melanie Alpaugh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.P.C.); (A.M.D.)
| |
Collapse
|
2
|
Vanalderwiert L, Henry A, Wahart A, Carvajal Berrio DA, Brauchle EM, El Kaakour L, Schenke-Layland K, Brinckmann J, Steenbock H, Debelle L, Six I, Faury G, Jaisson S, Gillery P, Durlach V, Sartelet H, Maurice P, Bennasroune A, Martiny L, Duca L, Romier B, Blaise S. Metabolic syndrome-associated murine aortic wall stiffening is associated with premature elastic fibers aging. Am J Physiol Cell Physiol 2024; 327:C698-C715. [PMID: 38946422 DOI: 10.1152/ajpcell.00615.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 07/02/2024]
Abstract
Type 2 diabetes (T2D) constitutes a major public health problem, and despite prevention efforts, this pandemic disease is one of the deadliest diseases in the world. In 2022, 6.7 million patients with T2D died prematurely from vascular complications. Indeed, diabetes increases the risk of myocardial infarction or stroke eightfold. The identification of the molecular factors involved in the occurrence of cardiovascular complications and their prevention are therefore major axes. Our hypothesis is that factors brought into play during physiological aging appear prematurely with diabetes progression. Our study focused on the aging of the extracellular matrix (ECM), a major element in the maintenance of vascular homeostasis. We characterized the morphological and functional aspects of aorta, with a focus on the collagen and elastic fibers of diabetic mice aged from 6 mo to nondiabetic mice aged 6 mo and 20 mo. The comparison with the two nondiabetic models (young and old) highlighted an exacerbated activity of proteases, which could explain a disturbance in the collagen accumulation and an excessive degradation of elastic fibers. Moreover, the generation of circulating elastin-derived peptides reflects premature aging of the ECM. These extracellular elements contribute to the appearance of vascular rigidity, often the origin of pathologies such as hypertension and atherosclerosis. In conclusion, we show that diabetic mice aged 6 mo present the same characteristics of ECM wear as those observed in mice aged 20 mo. This accelerated aortic wall remodeling could then explain the early onset of cardiovascular diseases and, therefore, the premature death of patients with T2D.NEW & NOTEWORTHY Aortic elastic fibers of young (6-mo old) individuals with diabetes degrade prematurely and exhibit an appearance like that found in aged (20-mo old) nondiabetic mice. Exacerbated elastolysis and elastin-derived peptide production are characteristic elements, contributing to early aortic wall rigidity and hypertension development. Therefore, limiting this early aging could be a judicious therapeutic approach to reduce cardiovascular complications and premature death in patients with diabetes.
Collapse
MESH Headings
- Animals
- Elastic Tissue/metabolism
- Elastic Tissue/pathology
- Vascular Stiffness/physiology
- Mice
- Aorta/metabolism
- Aorta/pathology
- Aorta/physiopathology
- Mice, Inbred C57BL
- Extracellular Matrix/metabolism
- Extracellular Matrix/pathology
- Male
- Metabolic Syndrome/metabolism
- Metabolic Syndrome/pathology
- Metabolic Syndrome/physiopathology
- Elastin/metabolism
- Collagen/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Aging/pathology
- Aging/metabolism
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Aging, Premature/metabolism
- Aging, Premature/pathology
- Aging, Premature/physiopathology
Collapse
Affiliation(s)
| | - Auberi Henry
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Amandine Wahart
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Daniel A Carvajal Berrio
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Eva M Brauchle
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute, Reutlingen, Germany
| | - Lara El Kaakour
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Katja Schenke-Layland
- Department for Medical Technologies and Regenerative Medicine, Institute of Biomedical Engineering, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute, Reutlingen, Germany
- Division of Cardiology, Department of Medicine/Cardiology, Cardiovascular Research Laboratories, David Geffen School of Medicine at UCLA, Los Angeles, California, United States
| | - Juergen Brinckmann
- Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany
| | - Heiko Steenbock
- Institute of Virology and Cell Biology, University of Lübeck, Lübeck, Germany
| | - Laurent Debelle
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Isabelle Six
- Research Unit 7517, Pathophysiological Mechanisms and Consequences of Cardiovascular Calcifications (MP3CV), University of Picardie Jules Verne, Amiens, France
| | - Gilles Faury
- University Grenoble Alpes, INSERM, CHU Grenoble Alpes, Grenoble, France
| | - Stéphane Jaisson
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
- Biochemistry Department, University Hospital of Reims, Reims, France
| | - Philippe Gillery
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
- Biochemistry Department, University Hospital of Reims, Reims, France
| | - Vincent Durlach
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
- Cardiovascular and Thoracic Division, Hôpital Robert Debré, Reims, France
| | - Hervé Sartelet
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Amar Bennasroune
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Laurent Martiny
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Béatrice Romier
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| | - Sébastien Blaise
- UMR CNRS 7369 MEDyc, University of Reims Champagne-Ardenne, Reims, France
| |
Collapse
|
3
|
Chacon AN, Su W, Hou T, Guo Z, Gong MC. Exenatide administration time determines the effects on blood pressure dipping in db/db mice via modulation of food intake and sympathetic activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601700. [PMID: 39005289 PMCID: PMC11245019 DOI: 10.1101/2024.07.02.601700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Type 2 diabetics have an increased prevalence of hypertension and nondipping blood pressure (BP), which worsen cardiovascular outcomes. Exenatide, a short acting glucagon-like peptide-1 receptor agonist (GLP-1RA) used to treat type 2 diabetes, also demonstrates blood pressure (BP)-lowering effects. However, the mechanisms behind this and the impact of administration timing on BP dipping remain unclear. We investigated the effects of exenatide intraperitoneal injected at light onset (ZT0) or dark onset (ZT12) in diabetic (db/db) mice and nondiabetic controls. Using radio-telemetry and BioDAQ cages, we continuously monitored BP and food intake. Db/db mice exhibited non-dipping BP and increased food intake. ZT0 exenatide administration restored BP dipping by specifically lowering light-phase BP, while ZT12 exenatide reversed dipping by lowering dark-phase BP. These effects correlated with altered food intake patterns, and importantly, were abolished when food access was removed. Additionally, urinary norepinephrine excretion, measured by HPLC, was significantly reduced 6 hours post-exenatide at both ZT0 and ZT12, suggesting sympathetic nervous system involvement. Notably, combining exenatide with either ganglionic blocker mecamylamine or α-blocker prazosin did not enhance BP reduction beyond the individual effects of each blocker. These findings reveal that exenatide, when administered at light onset, restores BP dipping in db/db mice by suppressing light-phase food intake and sympathetic activity. Importantly, the efficacy of exenatide is dependent on food availability and its timing relative to circadian rhythms, highlighting the potential for chronotherapy in optimizing GLP-1RA- based treatments for type 2 diabetes and hypertension. Graphic Abstract Article Highlights Maintaining a normal blood pressure (BP) circadian rhythm is vital for cardiovascular health, but diabetes often disrupts this rhythm. The effect of exenatide, a GLP-1 receptor agonist (GLP-1RA), on BP rhythm in diabetes is uncertain.This study investigates the impact of exenatide administration timing on BP patterns in diabetic db/db mice.Findings indicate that exenatide given at the onset of rest restores normal BP dipping, while at the start of the active phase worsens BP rhythm by modulating food intake and sympathetic activity.Timing GLP-1 RA administration may optimize BP control and provide cardiovascular benefits for type 2 diabetes patients.
Collapse
|
4
|
Elshareif N, Gornick E, Gavini CK, Aubert G, Mansuy-Aubert V. Comparison of western diet-induced obesity and streptozotocin mouse models: insights into energy balance, somatosensory dysfunction, and cardiac autonomic neuropathy. Front Physiol 2023; 14:1238120. [PMID: 37885804 PMCID: PMC10598778 DOI: 10.3389/fphys.2023.1238120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
Metabolic disorders such as obesity and type 2 diabetes (T2D) are increasingly prevalent worldwide, necessitating a deeper comprehension of their underlying mechanisms. However, translating findings from animal research to human patients remains challenging. This study aimed to investigate the long-term effects of Streptozotocin (STZ) on metabolic, cardiac, and somatosensory function in mice fed a Western diet (WD) of high fat, sucrose, and cholesterol with low doses of STZ administration compared to mice fed WD alone. In our research, we thoroughly characterized energy balance and glucose homeostasis, as well as allodynia and cardiac function, all of which have been previously shown to be altered by WD feeding. Notably, our findings revealed that the treatment of WD-fed mice with STZ exacerbated dysfunction in glucose homeostasis via reduced insulin secretion in addition to impaired peripheral insulin signaling. Furthermore, both WD and WD + STZ mice exhibited the same degree of cardiac autonomic neuropathy, such as reduced heart rate variability and decreased protein levels of cardiac autonomic markers. Furthermore, both groups developed the same symptoms of neuropathic pain, accompanied by elevated levels of activating transcription factor 3 (Atf3) in the dorsal root ganglia. These discoveries enhance our understanding of metabolic activity, insulin resistance, neuropathy, and cardiac dysfunction of diet-induced models of obesity and diabetes. The exacerbation of impaired insulin signaling pathways by STZ did not lead to or worsen cardiac and somatosensory dysfunction. Additionally, they offer valuable insights into suitable diet induced translational mouse models, thereby advancing the development of potential interventions for associated conditions.
Collapse
Affiliation(s)
- Nadia Elshareif
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Emily Gornick
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Chaitanya K. Gavini
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gregory Aubert
- Division of Cardiology, Department of Internal Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Clinical Development, CSL Vifor, Glattbrugg, Switzerland
| | - Virginie Mansuy-Aubert
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
5
|
Sidhu SK, Aleman JO, Heffron SP. Obesity Duration and Cardiometabolic Disease. Arterioscler Thromb Vasc Biol 2023; 43:1764-1774. [PMID: 37650325 PMCID: PMC10544713 DOI: 10.1161/atvbaha.123.319023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
Cardiovascular disease risk is known to be influenced by both the severity of a risk factor and the duration of exposure (eg, LDL [low-density lipoprotein] cholesterol, tobacco smoke). However, this concept has been largely neglected within the obesity literature. While obesity severity has been closely linked with cardiometabolic diseases, the risk of developing these conditions among those with obesity may be augmented by greater obesity duration over the life span. Few longitudinal or contemporary studies have investigated the influence of both factors in combination-cumulative obesity exposure-instead generally focusing on obesity severity, often at a single time point, given ease of use and lack of established methods to encapsulate duration. Our review focuses on what is known about the influence of the duration of exposure to excess adiposity within the obesity-associated cardiometabolic disease risk equation by means of summarizing the hypothesized mechanisms for and evidence surrounding the relationships of obesity duration with diverse cardiovascular and metabolic disease. Through the synthesis of the currently available data, we aim to highlight the importance of a better understanding of the influence of obesity duration in cardiovascular and metabolic disease pathogenesis. We underscore the clinical importance of aggressive early attention to obesity identification and intervention to prevent the development of chronic diseases that arise from exposure to excess body weight.
Collapse
Affiliation(s)
- Sharnendra K. Sidhu
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Jose O. Aleman
- Laboratory of Translational Obesity Research, Division of Endocrinology, Diabetes & Metabolism, New York University Grossman School of Medicine, New York, NY, USA
| | - Sean P. Heffron
- Center for the Prevention of Cardiovascular Disease, Leon H. Charney Division of Cardiology, NYU Langone Health, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
6
|
Sharma A, Choi JSY, Watson AMD, Li L, Sonntag T, Lee MKS, Murphy AJ, De Blasio M, Head GA, Ritchie RH, de Haan JB. Cardiovascular characterisation of a novel mouse model that combines hypertension and diabetes co-morbidities. Sci Rep 2023; 13:8741. [PMID: 37253814 DOI: 10.1038/s41598-023-35680-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 05/22/2023] [Indexed: 06/01/2023] Open
Abstract
Epidemiologic data suggest that the prevalence of hypertension in patients with diabetes mellitus is ∼1.5-2.0 times greater than in matched non-diabetic patients. This co-existent disease burden exacerbates cardiac and vascular injury, leading to structural and functional changes to the myocardium, impaired cardiac function and heart failure. Oxidative stress and persistent low-grade inflammation underlie both conditions, and are identified as major contributors to pathological cardiac remodelling. There is an urgent need for effective therapies that specifically target oxidative stress and inflammation to protect against cardiac remodelling. Animal models are a valuable tool for testing emerging therapeutics, however, there is a notable lack of appropriate animal models of co-morbid diabetes and hypertension. In this study, we describe a novel preclinical mouse model combining diabetes and hypertension to investigate cardiac and vascular pathology of co-morbid disease. Type 1 diabetes was induced in spontaneously hypertensive, 8-week old, male Schlager (BPH/2) mice via 5 consecutive, daily injections of streptozotocin (55 mg/kg in citrate buffer; i.p.). Non-diabetic mice received citrate buffer only. After 10 weeks of diabetes induction, cardiac function was assessed by echocardiography prior to post-mortem evaluation of cardiomyocyte hypertrophy, interstitial fibrosis and inflammation by histology, RT-PCR and flow cytometry. We focussed on the oxidative and inflammatory stress pathways that contribute to cardiovascular remodelling. In particular, we demonstrate that markers of inflammation (monocyte chemoattractant protein; MCP-1), oxidative stress (urinary 8-isoprostanes) and fibrosis (connective tissue growth factor; CTGF) are significantly increased, whilst diastolic dysfunction, as indicated by prolonged isovolumic relaxation time (IVRT), is elevated in this diabetic and hypertensive mouse model. In summary, this pre-clinical mouse model provides researchers with a tool to test therapeutic strategies unique to co-morbid diabetes and hypertension, thereby facilitating the emergence of novel therapeutics to combat the cardiovascular consequences of these debilitating co-morbidities.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia.
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.
| | - Judy S Y Choi
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Anna M D Watson
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, Australia
| | - Leila Li
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Thomas Sonntag
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Man K S Lee
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Andrew J Murphy
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Miles De Blasio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Geoffrey A Head
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Rebecca H Ritchie
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Judy B de Haan
- Group Leader (Oxidative Stress Laboratory), Diabetic Complications Division, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Australia
- Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
7
|
Wang S, Li S, Zhai X, Rong P, He J, Liu L, He X, Liu W. Transcutaneous auricular vagal nerve stimulation releases extrapineal melatonin and reduces thermal hypersensitivity in Zucker diabetic fatty rats. Front Neurosci 2022; 16:916822. [PMID: 36033612 PMCID: PMC9403073 DOI: 10.3389/fnins.2022.916822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Type 2 diabetes (T2D) is the most common comorbidity of COVID-19, and both are related to the lack of circulating melatonin. In addition, chronic pain is a common sequela of both COVID-19 and T2D. Using a neuropathic pain model produced by sciatic nerve chronic constriction injury in Zucker diabetic fatty rats, a verified preclinical genetic T2D neuropathy animal model, this study aimed to show that transcutaneous auricular vagal nerve stimulation (taVNS) could elevate plasma melatonin concentration, upregulate the expression of melatonin receptors (MTRs) in the amygdala, and relieve peripheral neuropathic pain. Furthermore, taVNS would restore melatonin levels and relieve pain even in pinealectomized rats. On the contrary, intraperitoneally injected luzindole, a melatonin receptor antagonist, would attenuate the antinociceptive effects of taVNS. In conclusion, the mechanism of the therapeutic effect of taVNS on chronic pain involves the release of extrapineal melatonin and the positive regulation of the expression of central MTRs. This beneficial efficacy should be considered during COVID-19 rehabilitation in individuals with diabetes.
Collapse
Affiliation(s)
- Shuxing Wang
- School of Medicine, Foshan University, Foshan, China
- *Correspondence: Shuxing Wang,
| | - Shaoyuan Li
- Department of Physiology, Institute of Acupuncture and Moxibustion, Academy of Chinese Medical Sciences, Beijing, China
| | - Xu Zhai
- Department of Physiology, Institute of Acupuncture and Moxibustion, Academy of Chinese Medical Sciences, Beijing, China
| | - Peijing Rong
- Department of Physiology, Institute of Acupuncture and Moxibustion, Academy of Chinese Medical Sciences, Beijing, China
| | - Jietao He
- School of Medicine, Foshan University, Foshan, China
| | - Lina Liu
- School of Medicine, Foshan University, Foshan, China
| | - Xinxin He
- School of Medicine, Foshan University, Foshan, China
| | - Wenguo Liu
- School of Medicine, Foshan University, Foshan, China
| |
Collapse
|
8
|
Pesta D, Jordan J. INDY as a Therapeutic Target for Cardio-Metabolic Disease. Metabolites 2022; 12:metabo12030244. [PMID: 35323687 PMCID: PMC8949283 DOI: 10.3390/metabo12030244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
Decreased expression of the plasma membrane citrate transporter INDY (acronym I’m Not Dead, Yet) promotes longevity and protects from high-fat diet- and aging-induced metabolic derangements. Preventing citrate import into hepatocytes by different strategies can reduce hepatic triglyceride accumulation and improve hepatic insulin sensitivity, even in the absence of effects on body composition. These beneficial effects likely derive from decreased hepatic de novo fatty acid biosynthesis as a result of reduced cytoplasmic citrate levels. While in vivo and in vitro studies show that inhibition of INDY prevents intracellular lipid accumulation, body weight is not affected by organ-specific INDY inhibition. Besides these beneficial metabolic effects, INDY inhibition may also improve blood pressure control through sympathetic nervous system inhibition, partly via reduced peripheral catecholamine synthesis. These effects make INDY a promising candidate with bidirectional benefits for improving both metabolic disease and blood pressure control.
Collapse
Affiliation(s)
- Dominik Pesta
- German Aerospace Center (DLR), Institute of Aerospace Medicine, D-51147 Cologne, Germany;
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, D-50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, D-50931 Cologne, Germany
- Correspondence:
| | - Jens Jordan
- German Aerospace Center (DLR), Institute of Aerospace Medicine, D-51147 Cologne, Germany;
| |
Collapse
|
9
|
Wu Y, Chen L, Wang C. Understanding the molecular role of syndecan-1 in the regulation of caspase-6 during the progression of cardiac arrhythmia. Exp Ther Med 2021; 22:1180. [PMID: 34475970 PMCID: PMC8406766 DOI: 10.3892/etm.2021.10614] [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: 06/16/2020] [Accepted: 06/24/2021] [Indexed: 12/02/2022] Open
Abstract
The role of caspase-6 in heart disease is not well understood, particularly with respect to cardiac arrhythmia. Also, the function of syndecan-1 in the stimulation of inflammation or a regenerative response after cardiac injury is unclear. Leptin receptor-deficient (C57BL/KS-leprdb/leprdb) mice were used in the present study. In addition to developing type 2 diabetes, they also develop initial- and end-stage cardiac arrhythmia after 5 and 8 months, respectively. The initial and end-stage arrhythmias were confirmed through progressive variations in the PP intervals observable in electrocardiograms. Histopathological images of the cardiac tissue exhibited scattered and loosened cardiac cells at the initial stage of cardiac arrhythmia, whereas tissue hardness and extensive structural changes in cardiomyocytes were evident at the end stage. At the molecular level, the progressive upregulation of caspase-6 was observed as the cardiac arrhythmia progressed. In the initial stage of arrhythmia, immunohistochemistry revealed that caspase-6 was expressed at the surface of cardiac cells, suggesting that caspase-6 targeted the extracellular matrix, leading to a loosening of the cardiac tissue structure. In the end stage of cardiac arrhythmia, caspase-6 expression was abundant in the cytoplasm, as well as at the cell surface, suggesting that caspase-6 may have cleaved intermediate filaments, paving the way for cellular morphological changes and apoptosis. Notably, syndecan-1 was upregulated 5.8-fold in the initial stage of cardiac arrhythmia, but downregulated at the end stage. Syndecan-1 may restrict the expression of caspase-6 in the initial stage of cardiac arrhythmia, while its downregulation at the end stage may allow destructive changes via caspase-6 overexpression. Furthermore, the knockdown of syndecan-1 using small interfering RNA enhanced the expression of caspase-6 in the cardiac tissue by factors of 1.8 and 1.2 at the initial and end stages of cardiac arrhythmia, respectively, compared with that in non-silenced cardiac tissue. Therefore, it may be concluded that syndecan-1 plays a major role in the regulation of caspase-6 during the pathological stages of cardiac arrhythmia.
Collapse
Affiliation(s)
- Yuanchu Wu
- Department of Cardiology, Affiliated Wujiang Hospital of Nantong University, Suzhou, Jiangsu 215200, P.R. China
| | - Lin Chen
- Department of Cardiology, Affiliated Wujiang Hospital of Nantong University, Suzhou, Jiangsu 215200, P.R. China
| | - Caihong Wang
- Department of Cardiology, Affiliated Wujiang Hospital of Nantong University, Suzhou, Jiangsu 215200, P.R. China
| |
Collapse
|
10
|
Shin MK, Mitrut R, Gu C, Kim LJ, Yeung BH, Lee R, Pham L, Tang WY, Sham JSK, Cui H, Polotsky VY. Pharmacological and Genetic Blockade of Trpm7 in the Carotid Body Treats Obesity-Induced Hypertension. Hypertension 2021; 78:104-114. [PMID: 33993722 PMCID: PMC8192446 DOI: 10.1161/hypertensionaha.120.16527] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roxana Mitrut
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chenjuan Gu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bonnie H.Y. Yeung
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rachel Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luu Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wan-Yee Tang
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - James S. K. Sham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Hou T, Su W, Duncan MJ, Olga VA, Guo Z, Gong MC. Time-restricted feeding protects the blood pressure circadian rhythm in diabetic mice. Proc Natl Acad Sci U S A 2021; 118:e2015873118. [PMID: 34161259 PMCID: PMC8237651 DOI: 10.1073/pnas.2015873118] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The quantity and quality of food intake have been considered crucial for peoples' wellness. Only recently has it become appreciated that the timing of food intake is also critical. Nondipping blood pressure (BP) is prevalent in diabetic patients and is associated with increased cardiovascular events. However, the causes and mechanisms of nondipping BP in diabetes are not fully understood. Here, we report that food intake and BP were arrhythmic in diabetic db/db mice fed a normal chow diet ad libitum. Imposing a food intake diurnal rhythm by time-restricted feeding (TRF; food was only available for 8 h during the active phase) prevented db/db mice from developing nondipping BP and effectively restored the already disrupted BP circadian rhythm in db/db mice. Interestingly, increasing the time of food availability from 8 h to 12 h during the active dark phase in db/db mice prompted isocaloric feeding and still provided robust protection of the BP circadian rhythm in db/db mice. In contrast, neither 8-h nor 12-h TRF affected BP dipping in wild-type mice. Mechanistically, we demonstrate that TRF protects the BP circadian rhythm in db/db mice via suppressing the sympathetic activity during the light phase when they are inactive and fasting. Collectively, these data reveal a potentially pivotal role of the timing of food intake in the prevention and treatment of nondipping BP in diabetes.
Collapse
Affiliation(s)
- Tianfei Hou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Wen Su
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Marilyn J Duncan
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536
| | | | - Zhenheng Guo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536;
- Research and Development, Lexington Veterans Affairs Medical Center, Lexington, KY 40502
| | - Ming C Gong
- Department of Physiology, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
12
|
Obesity-associated hyperleptinemia alters the gliovascular interface of the hypothalamus to promote hypertension. Cell Metab 2021; 33:1155-1170.e10. [PMID: 33951475 PMCID: PMC8183500 DOI: 10.1016/j.cmet.2021.04.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/27/2021] [Accepted: 04/12/2021] [Indexed: 12/21/2022]
Abstract
Pathologies of the micro- and macrovascular systems are a hallmark of the metabolic syndrome, which can lead to chronically elevated blood pressure. However, the underlying pathomechanisms involved still need to be clarified. Here, we report that an obesity-associated increase in serum leptin triggers the select expansion of the micro-angioarchitecture in pre-autonomic brain centers that regulate hemodynamic homeostasis. By using a series of cell- and region-specific loss- and gain-of-function models, we show that this pathophysiological process depends on hypothalamic astroglial hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling downstream of leptin signaling. Importantly, several distinct models of HIF1α-VEGF pathway disruption in astrocytes are protected not only from obesity-induced hypothalamic angiopathy but also from sympathetic hyperactivity or arterial hypertension. These results suggest that hyperleptinemia promotes obesity-induced hypertension via a HIF1α-VEGF signaling cascade in hypothalamic astrocytes while establishing a novel mechanistic link that connects hypothalamic micro-angioarchitecture with control over systemic blood pressure.
Collapse
|
13
|
Huang C, AlMarabeh S, Cavers J, Abdulla MH, Johns EJ. Effects of intracerebroventricular leptin and orexin-A on the baroreflex control of renal sympathetic nerve activity in conscious rats fed a normal or high-fat diet. Clin Exp Pharmacol Physiol 2021; 48:585-596. [PMID: 33352624 DOI: 10.1111/1440-1681.13451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/21/2020] [Accepted: 12/01/2020] [Indexed: 11/30/2022]
Abstract
This study examined the effect of leptin and orexin-A on autonomic baroreflex control in conscious Wistar rats exposed to high-fat (45% fat) or normal (3.4%) diet for 4 weeks. Renal sympathetic nerve activity (RSNA), mean arterial pressure (MAP) and heart rate (HR) were monitored during the generation of baroreflex gain curves and acute volume expansion (VEP). Intracerebroventricular (ICV) leptin (1 μg/min) increased RSNA in the normal diet group (0.31 ± 0.04 vs 0.23 ± 0.03 mV/s) and MAP in the high-fat diet group (115 ± 5 vs 105 ± 5 mm Hg, P < .05). Orexin-A (50 ng/min) increased RSNA, HR and MAP in the high-fat diet group (0.26 ± 0.03 vs 0.22 ± 0.02 mV/s, 454 ± 8 vs 417 ± 12 beats/min, 117 ± 1 vs 108 ± 1 mm Hg) and the normal diet group (0.18 ± 0.05 vs 0.17 ± 0.05 mV/s, 465 ± 10 vs 426 ± 6 beats/min, 116 ± 2 vs 104 ± 3 mm Hg). Baroreflex sensitivity for RSNA was increased during ICV leptin by 50% in the normal diet group, compared to 14% in the high-fat diet group (P < .05). Similarly, orexin-A increased baroreflex sensitivity by 56% and 50% in the high-fat and normal diet groups, respectively (all P < .05). During ICV saline, VEP decreased RSNA by 31 ± 5% (P < .05) after 10 minutes and the magnitude of this response was blunted during ICV infusion of leptin (17 ± 2%, P < .05) but not orexin-A in the normal diet group. RSNA response to VEP was not changed during ICV leptin or orexin-A in the high-fat diet group. These findings indicate possible central roles for leptin and orexin-A in modulating the baroreflexes under normal or increased fat intake in conscious rats and potential therapeutic approaches for obesity associated hypertension.
Collapse
Affiliation(s)
- Chunlong Huang
- Department of Physiology, Western Gateway Building, University College Cork, Cork, Ireland
| | - Sara AlMarabeh
- Department of Physiology, Western Gateway Building, University College Cork, Cork, Ireland
| | - Jeremy Cavers
- Department of Physiology, Western Gateway Building, University College Cork, Cork, Ireland
| | - Mohammed H Abdulla
- Department of Physiology, Western Gateway Building, University College Cork, Cork, Ireland
| | - Edward J Johns
- Department of Physiology, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
14
|
Circadian variations of vasoconstriction and blood pressure in physiology and diabetes. Curr Opin Pharmacol 2021; 57:125-131. [PMID: 33721615 DOI: 10.1016/j.coph.2021.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 01/14/2023]
Abstract
The intrinsic vascular smooth muscle contraction and vasoconstriction show time-of-day variations, contributing to the blood pressure circadian rhythm, which is essential for cardiovascular health. This brief review provides an overview of our current understanding of the mechanisms underlying the time-of-day variations of vascular smooth muscle contraction. We discuss the potential contribution of the time-of-day variations of vasoconstriction to the physiological blood pressure circadian rhythm. Finally, we survey the data obtained in the type 2 diabetic db/db mouse model that demonstrate the alterations of the time-of-day variations of vasoconstriction and the nondipping blood pressure in diabetes.
Collapse
|
15
|
Willmes DM, Daniels M, Kurzbach A, Lieske S, Bechmann N, Schumann T, Henke C, El-Agroudy NN, Da Costa Goncalves AC, Peitzsch M, Hofmann A, Kanczkowski W, Kräker K, Müller DN, Morawietz H, Deussen A, Wagner M, El-Armouche A, Helfand SL, Bornstein SR, de Cabo R, Bernier M, Eisenhofer G, Tank J, Jordan J, Birkenfeld AL. The longevity gene mIndy (I'm Not Dead, Yet) affects blood pressure through sympathoadrenal mechanisms. JCI Insight 2021; 6:136083. [PMID: 33491666 PMCID: PMC7934862 DOI: 10.1172/jci.insight.136083] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Reduced expression of the plasma membrane citrate transporter INDY (acronym I’m Not Dead, Yet) extends life span in lower organisms. Deletion of the mammalian Indy (mIndy) gene in rodents improves metabolism via mechanisms akin to caloric restriction, known to lower blood pressure (BP) by sympathoadrenal inhibition. We hypothesized that mIndy deletion attenuates sympathoadrenal support of BP. Continuous arterial BP and heart rate (HR) were reduced in mINDY-KO mice. Concomitantly, urinary catecholamine content was lower, and the decreases in BP and HR by mIndy deletion were attenuated after autonomic ganglionic blockade. Catecholamine biosynthesis pathways were reduced in mINDY-KO adrenals using unbiased microarray analysis. Citrate, the main mINDY substrate, increased catecholamine content in pheochromocytoma cells, while pharmacological inhibition of citrate uptake blunted the effect. Our data suggest that deletion of mIndy reduces sympathoadrenal support of BP and HR by attenuating catecholamine biosynthesis. Deletion of mIndy recapitulates beneficial cardiovascular and metabolic responses to caloric restriction, making it an attractive therapeutic target. Deletion of mIndy reduces blood pressure and heart rate by attenuating catecholamine biosynthesis and recapitulates beneficial cardiovascular and metabolic responses to caloric restriction.
Collapse
Affiliation(s)
- Diana M Willmes
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Martin Daniels
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Department of Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany
| | - Anica Kurzbach
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Department of Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Stefanie Lieske
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and
| | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tina Schumann
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Christine Henke
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Nermeen N El-Agroudy
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | | | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Anja Hofmann
- Division of Vascular Endothelium and Microcirculation, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Waldemar Kanczkowski
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Kristin Kräker
- Experimental and Clinical Research Center, Max Delbruck Center for Molecular Medicine and Charité - University Hospital Berlin, Berlin, Germany
| | - Dominik N Müller
- Experimental and Clinical Research Center, Max Delbruck Center for Molecular Medicine and Charité - University Hospital Berlin, Berlin, Germany
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Andreas Deussen
- Department of Physiology, Medical Faculty Carl Gustav Carus, and
| | - Michael Wagner
- Department of Pharmacology and Toxicology, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Stephen L Helfand
- Department of Molecular Biology, Cell Biology & Biochemistry, Division of Biology and Medicine, Brown University, Providence, Rhode Island, USA
| | - Stephan R Bornstein
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, Maryland, USA
| | - Graeme Eisenhofer
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Institute of Clinical Chemistry and Laboratory Medicine, University Hospital and Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Jens Tank
- Aerospace Medicine, University of Cologne, Cologne, Germany
| | - Jens Jordan
- Aerospace Medicine, University of Cologne, Cologne, Germany.,Institute for Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Andreas L Birkenfeld
- Section of Metabolic and Vascular Medicine, Medical Clinic III, University Hospital and Medical Faculty Carl Gustav Carus and.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, Technical University Dresden, Dresden, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, University of Tübingen, Tübingen, Germany.,Department of Internal Medicine IV, Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany.,Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
16
|
Wang Z, Li H. Serum brain-derived neurotrophic factor levels in patients with diabetic neuropathic pain. Neurosci Lett 2021; 752:135655. [PMID: 33485990 DOI: 10.1016/j.neulet.2021.135655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/26/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Diabetic neuropathic pain (DNP) is one of the most common and severe complications in patients with diabetes. This study aimed to investigate serum brain-derived neurotrophic factor (BDNF) levels in patients with DNP and to evaluate the association between BDNF and disease severity. METHODS A total of 143 T2DM patients were included, according to clinical characteristics and douleur neuropathique 4 (DN4) questionnaire are divided into the DNP group (n = 78) and without the DNP group (n = 65). BDNF levels were measured by an enzyme-linked immunosorbent assay. Additionally, other biochemical characteristics were measured using routine laboratory methods. RESULTS Serum levels of BDNF was increased significantly in the DNP group compared to without DNP group. Meanwhile, a binary logistic regression model identified as revealed BDNF (OR = 1.178, 95 %CI = 1.064-1.305,p = 0.002) was a risk factor in T2DM patients. Furthermore, the serum BDNF levels positively correlated with VAS score in the DNP patients. CONCLUSIONS Serum BDNF was elevated in DNP patients and increased gradually with the VAS score. BDNF was identified as risk factors for pain in all T2DM patients.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
17
|
Lubberding AF, Pereira L, Xue J, Gottlieb LA, Matchkov VV, Gomez AM, Thomsen MB. Aberrant sinus node firing during β-adrenergic stimulation leads to cardiac arrhythmias in diabetic mice. Acta Physiol (Oxf) 2020; 229:e13444. [PMID: 31953990 DOI: 10.1111/apha.13444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/25/2019] [Accepted: 01/13/2020] [Indexed: 01/08/2023]
Abstract
AIM Cardiovascular complications, including cardiac arrhythmias, result in high morbidity and mortality in patients with type-2 diabetes mellitus (T2DM). Clinical and experimental data suggest electrophysiological impairment of the natural pacemaker of the diabetic heart. The present study examined sinoatrial node (SAN) arrhythmias in a mouse model of T2DM and physiologically probed their underlying cause. METHODS Electrocardiograms were obtained from conscious diabetic db/db and lean control db/+ mice. In vivo SAN function was probed through pharmacological autonomic modulation with isoprenaline, atropine and carbachol. Blood pressure stability and heart rate variability (HRV) were evaluated. Intrinsic SAN function was evaluated through ex vivo imaging of spontaneous Ca2+ transients in isolated SAN preparations. RESULTS While lean control mice showed constant RR intervals during isoprenaline challenge, the diabetic mice experienced SAN arrhythmias with large RR fluctuations in a dose-dependent manner. These arrhythmias were completely abolished by atropine pre-treatment, while carbachol pretreatment significantly increased SAN arrhythmia frequency in the diabetic mice. Blood pressure and HRV were comparable in db/db and db/+ mice, suggesting that neither augmented baroreceptor feedback nor autonomic neuropathy is a likely arrhythmia mechanism. Cycle length response to isoprenaline was comparable in isolated SAN preparations from db/db and db/+ mice; however, Ca2+ spark frequency was significantly increased in db/db mice compared to db/+ at baseline and after isoprenaline. CONCLUSION Our results demonstrate a dysfunction of cardiac pacemaking in an animal model of T2DM upon challenge with a β-adrenergic agonist. Ex vivo, higher Ca2+ spark frequency is present in diabetic mice, which may be directly linked to in vivo arrhythmias.
Collapse
Affiliation(s)
- Anniek F. Lubberding
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Laetitia Pereira
- Université Paris‐Saclay Inserm UMR‐S 1180 Châtenay‐Malabry France
| | - Jianbin Xue
- Université Paris‐Saclay Inserm UMR‐S 1180 Châtenay‐Malabry France
| | - Lisa A. Gottlieb
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | | | - Ana M. Gomez
- Université Paris‐Saclay Inserm UMR‐S 1180 Châtenay‐Malabry France
| | - Morten B. Thomsen
- Department of Biomedical Sciences Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
18
|
Hou T, Su W, Guo Z, Gong MC. A Novel Diabetic Mouse Model for Real-Time Monitoring of Clock Gene Oscillation and Blood Pressure Circadian Rhythm. J Biol Rhythms 2018; 34:51-68. [PMID: 30278816 DOI: 10.1177/0748730418803719] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Diabetic patients have an increased prevalence of blood pressure (BP) circadian rhythm disruption, which is associated with an increased risk of target organ damage and detrimental cardiovascular events. Limited information is available regarding the role of clock genes in the disruption of BP circadian rhythm in diabetes due to the lack of a diabetic animal model that allows real-time monitoring of clock gene oscillation. Here, we generated a novel diabetic db/db-mPer2Luc mouse model by crossing type 2 diabetic db/db mice with mPer2Luc knock-in mice. The daily rhythms of BP, heart rate, locomotor activity, and food and water intake were acquired by radiotelemetry or using metabolic chambers. The daily oscillation of mPer2 bioluminescence was recorded by LumiCycle in real-time in tissue explants and using the IVIS system in vivo. Our results show that db/db-mPer2Luc mice are obese, diabetic, and glucose intolerant. The db/db-mPer2Luc mice displayed a compromised BP daily rhythm, which was associated with disrupted daily rhythms in baroreflex sensitivity, locomotor activity, and metabolism, but not heart rate or food and water intake. The phase of the mPer2 daily oscillation was advanced to different extents in the explanted peripheral tissues from db/db-mPer2Luc mice relative to control mice. In contrast, no phase shift was detected in mPer2 daily oscillations in the explanted SCN. Moreover, advanced phase shift of the mPer2 daily oscillation was detected in the liver, kidney and submandibular gland in vivo of db/db-mPer2Luc mice. In conclusion, the diabetic db/db-mPer2Luc mouse is a novel animal model that allows real-time monitoring of mPer2 circadian rhythms ex vivo and in vivo. The results from db/db-mPer2Luc mice suggest that the desynchrony of mPer2 daily oscillation in peripheral tissues contributes to the loss of BP daily oscillation in diabetes.
Collapse
Affiliation(s)
- Tianfei Hou
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Wen Su
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Zhenheng Guo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky.,Research and Development, Lexington Veterans Affairs Medical Center, Lexington, Kentucky
| | - Ming C Gong
- Department of Physiology, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
19
|
Chaudhary P, Schreihofer AM. Improved glucose homeostasis in male obese Zucker rats coincides with enhanced baroreflexes and activation of the nucleus tractus solitarius. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1195-R1209. [PMID: 30256679 DOI: 10.1152/ajpregu.00195.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Young adult male obese Zucker rats (OZR) develop insulin resistance and hypertension with impaired baroreflex-mediated bradycardia and activation of nucleus tractus solitarius (NTS). Because type 1 diabetic rats also develop impaired baroreflex-mediated NTS activation, we hypothesized that improving glycemic control in OZR would enhance compromised baroreflexes and NTS activation. Fasting blood glucose measured by telemetry was comparable in OZR and lean Zucker rats (LZR) at 12-17 wk. However, with access to food, OZR were chronically hyperglycemic throughout this age range. By 15-17 wk of age, tail samples yielded higher glucose values than those measured by telemetry in OZR but not LZR, consistent with reports of exaggerated stress responses in OZR. Injection of glucose (1g/kg ip) produced larger rises in glucose and areas under the curve in OZR than LZR. Treatment with metformin (300 mg·kg-1·day-1) or pioglitazone (5 mg·kg-1·day-1) in drinking water for 2-3 wk normalized fed glucose levels in OZR with no effect in LZR. After metformin treatment, area under the curve for blood glucose after glucose injection was reduced in OZR and comparable to LZR. Hyperinsulinemia was slightly reduced by each treatment in OZR, but insulin was still greatly elevated compared with LZR. Neither treatment reduced hypertension in OZR, but both treatments significantly improved the blunted phenylephrine-induced bradycardia and NTS c-Fos expression in OZR with no effect in LZR. These data suggest that restoring glycemic control in OZR enhances baroreflex control of heart rate by improving the response of the NTS to raising arterial pressure, even in the presence of hyperinsulinemia and hypertension.
Collapse
Affiliation(s)
- Parul Chaudhary
- Department of Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, Texas
| | - Ann M Schreihofer
- Department of Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, Texas
| |
Collapse
|
20
|
Butcher JT, Mintz JD, Larion S, Qiu S, Ruan L, Fulton DJ, Stepp DW. Increased Muscle Mass Protects Against Hypertension and Renal Injury in Obesity. J Am Heart Assoc 2018; 7:e009358. [PMID: 30369309 PMCID: PMC6201396 DOI: 10.1161/jaha.118.009358] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 06/20/2018] [Indexed: 01/06/2023]
Abstract
Background Obesity compromises cardiometabolic function and is associated with hypertension and chronic kidney disease. Exercise ameliorates these conditions, even without weight loss. Although the mechanisms of exercise's benefits remain unclear, augmented lean body mass is a suspected mechanism. Myostatin is a potent negative regulator of skeletal muscle mass that is upregulated in obesity and downregulated with exercise. The current study tested the hypothesis that deletion of myostatin would increase muscle mass and reduce blood pressure and kidney injury in obesity. Methods and Results Myostatin knockout mice were crossed to db/db mice, and metabolic and cardiovascular functions were examined. Deletion of myostatin increased skeletal muscle mass by ≈50% to 60% without concomitant weight loss or reduction in fat mass. Increased blood pressure in obesity was prevented by the deletion of myostatin, but did not confer additional benefit against salt loading. Kidney injury was evident because of increased albuminuria, which was abolished in obese mice lacking myostatin. Glycosuria, total urine volume, and whole kidney NOX-4 levels were increased in obesity and prevented by myostatin deletion, arguing that increased muscle mass provides a multipronged defense against renal dysfunction in obese mice. Conclusions These experimental observations suggest that loss of muscle mass is a novel risk factor in obesity-derived cardiovascular dysfunction. Interventions that increase muscle mass, either through exercise or pharmacologically, may help limit cardiovascular disease in obese individuals.
Collapse
Affiliation(s)
- Joshua T. Butcher
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGA
| | - James D. Mintz
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGA
| | - Sebastian Larion
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGA
| | - Shuiqing Qiu
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGA
| | - Ling Ruan
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGA
| | - David J. Fulton
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGA
- Department of PharmacologyMedical College of Georgia at Augusta UniversityAugustaGA
| | - David W. Stepp
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGA
- Department of PhysiologyMedical College of Georgia at Augusta UniversityAugustaGA
| |
Collapse
|
21
|
Giri B, Dey S, Das T, Sarkar M, Banerjee J, Dash SK. Chronic hyperglycemia mediated physiological alteration and metabolic distortion leads to organ dysfunction, infection, cancer progression and other pathophysiological consequences: An update on glucose toxicity. Biomed Pharmacother 2018; 107:306-328. [PMID: 30098549 DOI: 10.1016/j.biopha.2018.07.157] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/15/2018] [Accepted: 07/31/2018] [Indexed: 02/09/2023] Open
Abstract
Chronic exposure of glucose rich environment creates several physiological and pathophysiological changes. There are several pathways by which hyperglycemia exacerbate its toxic effect on cells, tissues and organ systems. Hyperglycemia can induce oxidative stress, upsurge polyol pathway, activate protein kinase C (PKC), enhance hexosamine biosynthetic pathway (HBP), promote the formation of advanced glycation end-products (AGEs) and finally alters gene expressions. Prolonged hyperglycemic condition leads to severe diabetic condition by damaging the pancreatic β-cell and inducing insulin resistance. Numerous complications have been associated with diabetes, thus it has become a major health issue in the 21st century and has received serious attention. Dysregulation in the cardiovascular and reproductive systems along with nephropathy, retinopathy, neuropathy, diabetic foot ulcer may arise in the advanced stages of diabetes. High glucose level also encourages proliferation of cancer cells, development of osteoarthritis and potentiates a suitable environment for infections. This review culminates how elevated glucose level carries out its toxicity in cells, metabolic distortion along with organ dysfunction and elucidates the complications associated with chronic hyperglycemia.
Collapse
Affiliation(s)
- Biplab Giri
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India.
| | - Sananda Dey
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India; Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Tanaya Das
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Mrinmoy Sarkar
- Experimental Medicine and Stem Cell Research Laboratory, Department of Physiology, West Bengal State University, Barasat, Kolkata 700126, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Mokdumpur, Malda 732103, India.
| |
Collapse
|
22
|
Pires PW, McClain JL, Hayoz SF, Dorrance AM. Mineralocorticoid receptor antagonism prevents obesity-induced cerebral artery remodeling and reduces white matter injury in rats. Microcirculation 2018; 25:e12460. [PMID: 29758591 DOI: 10.1111/micc.12460] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/06/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Midlife obesity is a risk factor for dementia development. Obesity has also been linked to hyperaldosteronism, and this can be modeled in rats by high fat (HF) feeding from weaning. Aldosterone, or activation of the mineralocorticoid receptor (MR) causes cerebrovascular injury in lean hypertensive rats. We hypothesized that rats fed a HF diet would show inward middle cerebral artery (MCA) remodeling that could be prevented by MR antagonism. We further proposed that the cerebral artery remodeling would be associated with white mater injury. METHODS Three-week-old male Sprague-Dawley rats were fed a HF diet ± the MR antagonist canrenoic acid (Canr) for 17 weeks. Control rats received normal chow (control NC). MCA structure was assessed by pressure myography. RESULTS The MCAs from HF fed rats had smaller lumens and thicker walls when compared to arteries from control NC rats; Canr prevented the MCA remodeling associated with HF feeding. HF feeding increased the mRNA expression of markers of cell proliferation and vascular inflammation in cerebral arteries and Canr treatment prevented this. White mater injury was increased in the rats fed the HF diet and this was reduced by Canr treatment. The expression of doublecortin, a marker of new and immature neurons was reduced in HF fed rats, and MR antagonism normalized this. CONCLUSIONS These data suggest that HF feeding leads to MR dependent remodeling of the MCA and this is associated with markers of dementia development.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Department of Pharmacology, Center for Cardiovascular Research, University of Nevada School of Medicine, Reno, NV, USA
| | - Jonathon L McClain
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Sebastian F Hayoz
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.,Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN, USA
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
23
|
Tsuneki H, Wada T, Sasaoka T. Chronopathophysiological implications of orexin in sleep disturbances and lifestyle-related disorders. Pharmacol Ther 2018; 186:25-44. [DOI: 10.1016/j.pharmthera.2017.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
24
|
Han X, Ross J, Kolumam G, Pi M, Sonoda J, King G, Quarles LD. Cardiovascular Effects of Renal Distal Tubule Deletion of the FGF Receptor 1 Gene. J Am Soc Nephrol 2018; 29:69-80. [PMID: 28993502 PMCID: PMC5748915 DOI: 10.1681/asn.2017040412] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/03/2017] [Indexed: 01/11/2023] Open
Abstract
The bone-derived hormone fibroblast growth factor-23 (FGF-23) activates complexes composed of FGF receptors (FGFRs), including FGFR1, and α-Klotho in the kidney distal tubule (DT), leading to increased sodium retention and hypertension. However, the role of FGFR1 in regulating renal processes linked to hypertension is unclear. Here, we investigated the effects of selective FGFR1 loss in the DT. Conditional knockout (cKO) of FGFR1 in the DT (FGFR1DT-cKO mice) resulted in left ventricular hypertrophy (LVH) and decreased kidney expression of α-Klotho in association with enhanced BP, decreased expression of angiotensin converting enzyme 2, and increased expression of the Na+-K+-2Cl- cotransporter. Notably, recombinant FGF-23 administration similarly decreased the kidney expression of α-Klotho and induced LVH in mice. Pharmacologic activation of FGFR1 with a monoclonal anti-FGFR1 antibody (R1MAb1) normalized BP and significantly attenuated LVH in the Hyp mouse model of excess FGF-23, but did not induce a response in FGFR1DT-cKO mice. The hearts of FGFR1DT-cKO mice showed increased expression of the transient receptor potential cation channel, subfamily C, member 6 (TRPC6), consistent with cardiac effects of soluble Klotho deficiency. Moreover, administration of recombinant soluble Klotho lowered BP in the Hyp mice. Thus, FGFR1 in the DT regulates systemic hemodynamic responses opposite to those predicted by the actions of FGF-23. These cardiovascular effects appear to be mediated by paracrine FGF control of kidney FGFR1 and subsequent regulation of soluble Klotho and TRPC6. FGFR1 in the kidney may provide a new molecular target for treating hypertension.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2
- Animals
- Antibodies, Monoclonal/pharmacology
- Blood Pressure/drug effects
- Blood Pressure/genetics
- Female
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/pharmacology
- Glucuronidase/genetics
- Glucuronidase/metabolism
- Hypertension/genetics
- Hypertrophy, Left Ventricular/genetics
- Immunologic Factors/pharmacology
- Kidney Tubules, Distal
- Klotho Proteins
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/metabolism
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- RNA, Messenger/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/immunology
- Recombinant Proteins/pharmacology
- Sodium-Potassium-Chloride Symporters/genetics
- Sodium-Potassium-Chloride Symporters/metabolism
- TRPC Cation Channels/genetics
- TRPC Cation Channels/metabolism
- TRPC6 Cation Channel
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jed Ross
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Ganesh Kolumam
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Min Pi
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Junichiro Sonoda
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Gwendalyn King
- Department of Neurobiology, University of Alabama in Birmingham, Birmingham, Alabama
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee;
| |
Collapse
|
25
|
Rodgers JL, Samal E, Mohapatra S, Panguluri SK. Hyperoxia-induced cardiotoxicity and ventricular remodeling in type-II diabetes mice. Heart Vessels 2017; 33:561-572. [PMID: 29209776 DOI: 10.1007/s00380-017-1100-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/01/2017] [Indexed: 12/14/2022]
Abstract
Hyperoxia, or supplemental oxygen, is regularly used in the clinical setting for critically ill patients in ICU. However, several recent studies have demonstrated the negative impact of this treatment in patients in critical care, including increased rates of lung and cardiac injury, as well as increased mortality. The purpose of this study was to determine the predisposition for arrhythmias and electrical remodeling in a type 2 diabetic mouse model (db/db), as a result of hyperoxia treatment. For this, db/db and their heterozygous controls were treated with hyperoxia (> 90% oxygen) or normoxia (normal air) for 72-h. Immediately following hyperoxia or normoxia treatments, mice underwent surface ECG. Excised left ventricles were used to assess ion channel expression, including for Kv1.4, Kv1.5, Kv4.2, and KChIP2. Serum cardiac markers were also measured, including cardiac troponin I and lactate dehydrogenase. Our results showed that db/db mice have increased sensitivity to arrhythmia. Normoxia-treated db/db mice displayed features of arrhythmia, including QTc and JT prolongation, as well as QRS prolongation. A significant increase in QRS prolongation was also observed in hyperoxia-treated db/db mice, when compared to hyperoxia-treated heterozygous control mice. Db/db mice were also shown to exhibit ion channel dysregulation, as demonstrated by down-regulation in Kv1.5, Kv4.2, and KChIP2 under hyperoxia conditions. From these results, we conclude that: (1) diabetic mice showed distinct pathophysiology, when compared to heterozygous controls, both in normoxia and hyperoxia conditions. (2) Diabetic mice were more susceptible to arrhythmia at normal air conditions; this effect was exacerbated at hyperoxia conditions. (3) Unlike in heterozygous controls, diabetic mice did not demonstrate cardiac hypertrophy as a result of hyperoxia. (4) Ion channel remodeling was also observed in db/db mice under hyperoxia condition similar to its heterozygous controls.
Collapse
Affiliation(s)
- Jennifer Leigh Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., MDC-30, Tampa, FL, 33612, USA
| | - Eva Samal
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Siva Kumar Panguluri
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B. Downs Blvd., MDC-30, Tampa, FL, 33612, USA.
| |
Collapse
|
26
|
Xie Y, Potter CMF, Le Bras A, Nowak WN, Gu W, Bhaloo SI, Zhang Z, Hu Y, Zhang L, Xu Q. Leptin Induces Sca-1 + Progenitor Cell Migration Enhancing Neointimal Lesions in Vessel-Injury Mouse Models. Arterioscler Thromb Vasc Biol 2017; 37:2114-2127. [PMID: 28935755 PMCID: PMC5671780 DOI: 10.1161/atvbaha.117.309852] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/08/2017] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Leptin is an adipokine initially thought to be a metabolic factor. Recent publications have shown its roles in inflammation and vascular disease, to which Sca-1+ vascular progenitor cells within the vessel wall may contribute. We sought to elucidate the effects of leptin on Sca-1+ progenitor cells migration and neointimal formation and to understand the underlying mechanisms. Approach and Results— Sca-1+ progenitor cells from the vessel wall of Lepr+/+ and Lepr−/− mice were cultured and purified. The migration of Lepr+/+ Sca-1+ progenitor cells in vitro was markedly induced by leptin. Western blotting and kinase assays revealed that leptin induced the activation of phosphorylated signal transducer and activator of transcription 3, phosphorylated extracellular signal–regulated kinases 1/2, pFAK (phosphorylated focal adhesion kinase), and Rac1 (ras-related C3 botulinum toxin substrate 1)/Cdc42 (cell division control protein 42 homolog). In a mouse femoral artery guidewire injury model, an increased expression of leptin in both injured vessels and serum was observed 24 hours post-surgery. RFP (red fluorescent protein)-Sca-1+ progenitor cells in Matrigel were applied to the adventitia of the injured femoral artery. RFP+ cells were observed in the intima 24 hours post-surgery, subsequently increasing neointimal lesions at 2 weeks when compared with the arteries without seeded cells. This increase was reduced by pre-treatment of Sca-1+ cells with a leptin antagonist. Guidewire injury could only induce minor neointima in Lepr−/− mice 2 weeks post-surgery. However, transplantation of Lepr+/+ Sca-1+ progenitor cells into the adventitial side of injured artery in Lepr−/− mice significantly enhanced neointimal formation. Conclusions— Upregulation of leptin levels in both the vessel wall and the circulation after vessel injury promoted the migration of Sca-1+ progenitor cells via leptin receptor–dependent signal transducer and activator of transcription 3- Rac1/Cdc42-ERK (extracellular signal–regulated kinase)-FAK pathways, which enhanced neointimal formation.
Collapse
Affiliation(s)
- Yao Xie
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Claire M F Potter
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Alexandra Le Bras
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Witold N Nowak
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Wenduo Gu
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Shirin Issa Bhaloo
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Zhongyi Zhang
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Yanhua Hu
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Li Zhang
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.)
| | - Qingbo Xu
- From the Cardiovascular Division, King's College London BHF Centre, United Kingdom (Y.X., C.M.F.P., W.N.N., A.L.B., W.G., S.I.B., Z.Z., Y.H., Q.X.); and Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (L.Z.).
| |
Collapse
|
27
|
Lanfray D, Richard D. Emerging Signaling Pathway in Arcuate Feeding-Related Neurons: Role of the Acbd7. Front Neurosci 2017; 11:328. [PMID: 28690493 PMCID: PMC5481368 DOI: 10.3389/fnins.2017.00328] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/24/2017] [Indexed: 01/28/2023] Open
Abstract
The understanding of the mechanisms whereby energy balance is regulated is essential to the unraveling of the pathophysiology of obesity. In the last three decades, focus was put on the metabolic role played by the hypothalamic neurons expressing proopiomelanocortin (POMC) and cocaine and amphetamine regulated transcript (CART) and the neurons co-localizing agouti-related peptide (AgRP), neuropeptide Y (NPY), and gamma-aminobutyric acid (GABA). These neurons are part of the leptin-melanocortin pathway, whose role is key in energy balance regulation. More recently, the metabolic involvement of further hypothalamic uncharacterized neuron populations has been suggested. In this review, we discuss the potential homeostatic implication of hypothalamic GABAergic neurons that produce Acyl-Coa-binding domain containing protein 7 (ACBD7), precursor of the nonadecaneuropeptide (NDN), which has recently been characterized as a potent anorexigenic neuropeptide capable of relaying the leptin anorectic/thermogenic effect via the melanocortin system.
Collapse
Affiliation(s)
- Damien Lanfray
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université LavalQuébec, QC, Canada
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université LavalQuébec, QC, Canada
| |
Collapse
|
28
|
Sensory and autonomic function and structure in footpads of a diabetic mouse model. Sci Rep 2017; 7:41401. [PMID: 28128284 PMCID: PMC5269750 DOI: 10.1038/srep41401] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022] Open
Abstract
Sensory and autonomic neuropathy affects the majority of type II diabetic patients. Clinically, autonomic evaluation often focuses on sudomotor function yet this is rarely assessed in animal models. We undertook morphological and functional studies to assess large myelinated and small unmyelinated axons in the db/db type II diabetes mouse model. We observed that autonomic innervation of sweat glands in the footpads was significantly reduced in db/db mice compared to control db/+ mice and this deficit was greater compared to reductions in intraepidermal sensory innervation of adjacent epidermis. Additionally, db/db mice formed significantly fewer sweat droplets compared to controls as early as 6 weeks of age, a time when no statistical differences were observed electrophysiologically between db/db and db/+ mice studies of large myelinated sensory and motor nerves. The rate of sweat droplet formation was significantly slower and the sweat droplet size larger and more variable in db/db mice compared to controls. Whereas pilocarpine and glycopyrrolate increased and decreased sweating, respectively, in 6 month-old controls, db/db mice did not respond to pharmacologic manipulations. Our findings indicate autonomic neuropathy is an early and prominent deficit in the db/db model and have implications for the development of therapies for peripheral diabetic neuropathy.
Collapse
|
29
|
Habibi J, Aroor AR, Sowers JR, Jia G, Hayden MR, Garro M, Barron B, Mayoux E, Rector RS, Whaley-Connell A, DeMarco VG. Sodium glucose transporter 2 (SGLT2) inhibition with empagliflozin improves cardiac diastolic function in a female rodent model of diabetes. Cardiovasc Diabetol 2017; 16:9. [PMID: 28086951 PMCID: PMC5237274 DOI: 10.1186/s12933-016-0489-z] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/25/2016] [Indexed: 01/08/2023] Open
Abstract
Obese and diabetic individuals are at increased risk for impairments in diastolic relaxation and heart failure with preserved ejection fraction. The impairments in diastolic relaxation are especially pronounced in obese and diabetic women and predict future cardiovascular disease (CVD) events in this population. Recent clinical data suggest sodium glucose transporter-2 (SGLT2) inhibition reduces CVD events in diabetic individuals, but the mechanisms of this CVD protection are unknown. To determine whether targeting SGLT2 improves diastolic relaxation, we utilized empagliflozin (EMPA) in female db/db mice. Eleven week old female db/db mice were fed normal mouse chow, with or without EMPA, for 5 weeks. Blood pressure (BP), HbA1c and fasting glucose were significantly increased in untreated db/db mice (DbC) (P < 0.01). EMPA treatment (DbE) improved glycemic indices (P < 0.05), but not BP (P > 0.05). At baseline, DbC and DbE had already established impaired diastolic relaxation as indicated by impaired septal wall motion (>tissue Doppler derived E'/A' ratio) and increased left ventricular (LV) filling pressure (<E/E' ratio). Although these abnormalities persisted throughout the study period in DbC, diastolic function improved with EMPA treatment. In DbC, myocardial fibrosis was accompanied by increased expression of profibrotic/prohypertrophic proteins, serum/glucocorticoid regulated kinase 1 (SGK1) and the epithelial sodium channel (ENaC), and the development of these abnormalities were reduced with EMPA. DbC exhibited eccentric LV hypertrophy that was slightly improved by EMPA, indicated by a reduction in cardiomyocyte cross sectional area. In summary, EMPA improved glycemic indices along with diastolic relaxation, as well as SGK1/ENaC profibrosis signaling and associated interstitial fibrosis, all of which occurred in the absence of any changes in BP.
Collapse
Affiliation(s)
- Javad Habibi
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Annayya R Aroor
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - James R Sowers
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA.,Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine, Columbia, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,The Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Guanghong Jia
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Melvin R Hayden
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA
| | - Mona Garro
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Brady Barron
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Eric Mayoux
- Department of Cardiometabolic Diseases Research, Boehringer-Ingelheim, Biberach, Germany
| | - R Scott Rector
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Departments of Medicine-Gastroenterology and Hepatology and Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Adam Whaley-Connell
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA.,Division of Nephrology, University of Missouri, School of Medicine, Columbia, USA.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Vincent G DeMarco
- Department of Medicine, Division of Endocrinology, Diabetes and Cardiovascular Center, University of Missouri, School of Medicine, Columbia, USA. .,Department of Medical Pharmacology and Physiology, University of Missouri, School of Medicine, Columbia, USA. .,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.
| |
Collapse
|
30
|
Tsuneki H, Kon K, Ito H, Yamazaki M, Takahara S, Toyooka N, Ishii Y, Sasahara M, Wada T, Yanagisawa M, Sakurai T, Sasaoka T. Timed Inhibition of Orexin System by Suvorexant Improved Sleep and Glucose Metabolism in Type 2 Diabetic db/db Mice. Endocrinology 2016; 157:4146-4157. [PMID: 27631554 DOI: 10.1210/en.2016-1404] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Sleep disturbances are associated with type 2 diabetes; therefore, the amelioration of sleep may improve metabolic disorders. To investigate this possibility, we here examined the effects of suvorexant, an antiinsomnia drug targeting the orexin system, on sleep and glucose metabolism in type 2 diabetic mice. Diabetic db/db mice had a longer wakefulness time during the resting period, as compared with nondiabetic db/m+ control mice. The single or 7-day administration of suvorexant at lights-on (ie, the beginning of the resting phase) increased nonrapid eye movement sleep time during the resting phase and, as a consequence, reduced awake time. The daily resting-phase administration of suvorexant for 2-4 weeks improved impaired glucose tolerance in db/db mice without affecting body weight gain, food intake, systemic insulin sensitivity, or serum insulin, and glucagon levels. No changes were detected in the markers of lipid metabolism and inflammation, such as the hepatic triglyceride content and Tnf-α mRNA levels in liver and adipose tissues. The improving effect of suvorexant on glucose tolerance was associated with a reduction in the expression levels of hepatic gluconeogenic factors, including phosphoenolpyruvate carboxykinase and peroxisome proliferator-activated receptor-γ coactivator-1α in the liver in the resting phase. In contrast, the daily awake-phase administration of suvorexant had no beneficial effect on glucose metabolism. These results suggest that the suvorexant-induced increase of sleep time at the resting phase improved hepatic glucose metabolism in db/db mice. Our results provide insight into the development of novel pharmacological interventions for type 2 diabetes that target the orexin-operated sleep/wake regulatory system.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Kanta Kon
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Hisakatsu Ito
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Mitsuaki Yamazaki
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Satoyuki Takahara
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Naoki Toyooka
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Yoko Ishii
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Masakiyo Sasahara
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Tsutomu Wada
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Masashi Yanagisawa
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Takeshi Sakurai
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| | - Toshiyasu Sasaoka
- Departments of Clinical Pharmacology (H.T., K.K., T.W., T.Sas.) and Anesthesiology (H.I., M.Yam.), Graduate School of Science and Technology, and Graduate School of Innovative Life Science (S.T., N.T.), and Department of Pathology (Y.I., M.S.), University of Toyama, Toyama 930-0194, Japan; and International Institute for Integrative Sleep Medicine (WPI-IIIS) (M.Yan., T.Sak.), University of Tsukuba, Tsukuba 305-8575, Japan
| |
Collapse
|
31
|
Tsuneki H, Sasaoka T, Sakurai T. Sleep Control, GPCRs, and Glucose Metabolism. Trends Endocrinol Metab 2016; 27:633-642. [PMID: 27461005 DOI: 10.1016/j.tem.2016.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/29/2022]
Abstract
Modern lifestyles prolong daily activities into the nighttime, disrupting circadian rhythms, which may cause sleep disturbances. Sleep disturbances have been implicated in the dysregulation of blood glucose levels and reported to increase the risk of type 2 diabetes (T2D) and diabetic complications. Sleep disorders are treated using anti-insomnia drugs that target ionotropic and G protein-coupled receptors (GPCRs), including γ-aminobutyric acid (GABA) agonists, melatonin agonists, and orexin receptor antagonists. A deeper understanding of the effects of these medications on glucose metabolism and their underlying mechanisms of action is crucial for the treatment of diabetic patients with sleep disorders. In this review we focus on the beneficial impact of sleep on glucose metabolism and suggest a possible strategy for therapeutic intervention against sleep-related metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Takeshi Sakurai
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
32
|
Miranda-Anaya M, Carmona-Alcocer V, Carmona-Castro A. Effects of obesity on circadian photic entrainment of locomotor activity in wild miceNeotomodon alstoni. BIOL RHYTHM RES 2016. [DOI: 10.1080/09291016.2016.1158906] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Abstract
Abdominal obesity and elevated blood pressure commonly occur in the same patient and are key components of the metabolic syndrome. However, the association between obesity and increased blood pressure is variable. We review mechanisms linking cardiovascular and metabolic disease in such patients including altered systemic and regional hemodynamic control, neurohumoral activation, and relative natriuretic peptide deficiency. Moreover, we discuss recent results using omics techniques providing insight in molecular pathways linking adiposity, metabolic disease, and arterial hypertension. Recognition of the mechanisms orchestrating the crosstalk between cardiovascular and metabolic regulation in individual patients may lead to better and more precise treatments. It is reassuring that recently developed cardiovascular and metabolic medications may in fact ameliorate, both, cardiovascular and metabolic risks.
Collapse
Affiliation(s)
- Jens Jordan
- Institute for Clinical Pharmacology, Medical School Hannover, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | - Andreas L Birkenfeld
- Section of Metabolic Vascular Medicine, Medical Clinic III, Dresden University School of Medicine, Dresden, TU, Germany
- Center for Clinical Studies, GWT-TUD GmbH, Dresden, Germany
- Paul Langerhans Institute Dresden (PLID), A Member of the German Center for Diabetes Research (DZD e.V.), Dresden, Germany
| |
Collapse
|
34
|
Langer S, Beescho C, Ring A, Dorfmann O, Steinau HU, Spindler N. A new in vivo model using a dorsal skinfold chamber to investigate microcirculation and angiogenesis in diabetic wounds. GMS INTERDISCIPLINARY PLASTIC AND RECONSTRUCTIVE SURGERY DGPW 2016; 5:Doc09. [PMID: 26955508 PMCID: PMC4764794 DOI: 10.3205/iprs000088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Introduction: Diabetes mellitus describes a dysregulation of glucose metabolism due to improper insulin secretion, reduced insulin efficacy or both. It is a well-known fact that diabetic patients are likely to suffer from impaired wound healing, as diabetes strongly affects tissue angiogenesis. Until now, no satisfying in vivo murine model has been established to analyze the dynamics of angiogenesis during diabetic wound healing. To help understand the pathophysiology of diabetes and its effect on angiogenesis, a novel in vivo murine model was established using the skinfold chamber in mice. Materials and Methods: Mutant diabetic mice (db; BKS.Cg-m+/+Leprdb/J), wildtype mice (dock7Leprdb+/+m) and laboratory BALB/c mice were examined. They were kept in single cages with access to laboratory chow with an 12/12 hour day/night circle. Lesions of the panniculus muscle (Ø 2 mm) were created in the center of the transparent window chamber and the subsequent muscular wound healing was then observed for a period of 22 days. Important analytic parameters included vessel diameter, red blood cell velocity, vascular permeability, and leakage of muscle capillaries and post capillary venules. The key parameters were functional capillary density (FCD) and angiogenesis positive area (APA). Results: We established a model which allows high resolution in vivo imaging of functional angiogenesis in diabetic wounds. As expected, db mice showed impaired wound closure (day 22) compared to wounds of BALB/c or WT mice (day 15). FCD was lower in diabetic mice compared to WT and BALB/c during the entire observation period. The dynamics of angiogenesis also decreased in db mice, as reflected by the lowest APA levels. Significant variations in the skin buildup were observed, with the greatest skin depth in db mice. Furthermore, in db mice, the dermis:subcutaneous ratio was highly shifted towards the subcutaneous layers as opposed to WT or BALB/c mice. Conclusion: Using this new in vivo model of the skinfold chamber, it was possible to analyze and quantify microangiopathical changes which are essential for a better understanding of the pathophysiology of disturbed wound healing. Research in microcirculation is important to display perfusion in wounds versus healthy tissue. Using our model, we were able to compare wound healing in diabetic and healthy mice. We were also able to objectively analyze perfusion in wound edges and compare microcirculatory parameters. This model may be well suited to augment different therapeutic options.
Collapse
Affiliation(s)
- Stefan Langer
- Department of Plastic, Esthetic and Special Hand Surgery, University Hospital Leipzig, Germany
| | - Christian Beescho
- Department of Plastic, Esthetic and Special Hand Surgery, University Hospital Leipzig, Germany
| | - Andrej Ring
- Department of Plastic Surgery and Severe Burns, University Hospital Bergmannsheil, Ruhr University Bochum, Germany
| | - Olivia Dorfmann
- Department of Plastic, Esthetic and Special Hand Surgery, University Hospital Leipzig, Germany
| | | | - Nick Spindler
- Department of Plastic, Esthetic and Special Hand Surgery, University Hospital Leipzig, Germany
| |
Collapse
|
35
|
Differential impact of type-1 and type-2 diabetes on control of heart rate in mice. Auton Neurosci 2015; 194:17-25. [PMID: 26725752 DOI: 10.1016/j.autneu.2015.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 11/23/2015] [Accepted: 12/14/2015] [Indexed: 01/14/2023]
Abstract
AIMS Cardiac autonomic dysfunction is a serious complication of diabetes. One consequence is disruption of the normal beat-to-beat regulation of heart rate (HR), i.e. HR variability (HRV). However, our understanding of the disease process has been limited by inconsistent HR/HRV data from previous animal studies. We hypothesized that differences in the method of measurement, time of day, and level of stress account for the differing results across studies. Thus, our aim was to systematically assess HR and HRV in two common diabetic mouse models. METHODS ECG radiotelemetry devices were implanted into db/db (type-2 diabetic), STZ-treated db/+ (type-1 diabetic), and control db/+ mice (n=4 per group). HR and HRV were analyzed over 24 h and during treadmill testing. RESULTS 24 h analysis revealed that db/db mice had an altered pattern of circadian HR changes, and STZ-treated mice had reduced HR throughout. HRV measures linked to sympathetic control were reduced in db/db mice in the early morning and early afternoon, and partially reduced in STZ-treated mice. HR response to treadmill testing was blunted in both models. CONCLUSIONS It is important to consider both time of day and level of stress when assessing HR and HRV in diabetic mice. db/db mice may have altered circadian rhythm of sympathetic control of HR, whereas STZ-treated mice have a relative reduction. This study provides baseline data and a framework for HR analysis that may guide future investigations.
Collapse
|
36
|
Zhai X, Sun C, Rong P, Li S, McCabe MF, Wang X, Mao J, Wang S. A Correlative Relationship Between Chronic Pain and Insulin Resistance in Zucker Fatty Rats: Role of Downregulation of Insulin Receptors. THE JOURNAL OF PAIN 2015; 17:404-13. [PMID: 26705975 DOI: 10.1016/j.jpain.2015.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/29/2015] [Accepted: 12/07/2015] [Indexed: 02/08/2023]
Abstract
UNLABELLED Epidemiological studies and meta-analyses report a strong relationship between chronic pain and abnormalities in glucose metabolism, but the exact relationship between chronic pain and insulin resistance in type 2 diabetes (T2D) remains unknown. Using a model of neuropathic thermal and tactile hypersensitivity induced by chronic constriction injury (CCI) of the sciatic nerve in Zucker Diabetic Fatty (ZDF) and Zucker Lean (ZL) littermates, we compared the recovery period of hypersensitivity and the progression of T2D and studied the possible involvement of insulin receptors (IRs) in the comorbidity of these 2 conditions. We found that the nociceptive thresholds to thermal and mechanical stimulation in naive ZDF rats were lower than in ZL littermates at 6 weeks of age. Although ZDF and ZL rats developed thermal and tactile hypersensitivity after CCI, it took a longer time nociceptive sensitivity to be restored in ZDF rats. Nerve injury accelerated the progression of T2D in ZDF rats, shown by an earlier onset of hyperglycemia, more severe hyperinsulinemia, and a higher concentration of glycosylated hemoglobin Alc 6 weeks after CCI, compared with those in naive ZDF and ZL rats. IR-immunoreactive cells were located across the central nervous system and skeletal muscles. In the central nervous system, IR coexpressed with a neuronal marker (neuronal nuclei) but not a glial marker (glial fibrillary acidic protein). There was a low level of IR expression in skeletal muscles of naive ZDF rats. In contrast, CCI reduced the IR expression in skeletal muscles as well as the ipsilateral spinal cord, primarily in the dorsal horn. In conclusion, our data suggest that the relationship between insulin resistance and chronic pain in ZDF rats is bidirectional and an impaired IR signaling system might be implicated in this reciprocal relationship. PERSPECTIVE Nerve injuries in genetically susceptible individuals might accelerate the development of insulin resistance as in T2D. A downregulated expression of IRs in the skeletal muscle innervated by the injured nerve is one of the underlying mechanisms.
Collapse
Affiliation(s)
- Xu Zhai
- Department of Anatomy, Xinxiang Medical University, Xinxiang, Henan Province, China; Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunli Sun
- Department of Anatomy, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Peijing Rong
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shaoyuan Li
- Department of Physiology, Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Michael F McCabe
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xing Wang
- Department of Anatomy, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Jianren Mao
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shuxing Wang
- Guangdong Institute of Applied Biological Resources, Guangzhou, China; Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Guangzhou, China.
| |
Collapse
|
37
|
Labbé SM, Caron A, Lanfray D, Monge-Rofarello B, Bartness TJ, Richard D. Hypothalamic control of brown adipose tissue thermogenesis. Front Syst Neurosci 2015; 9:150. [PMID: 26578907 PMCID: PMC4630288 DOI: 10.3389/fnsys.2015.00150] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022] Open
Abstract
It has long been known, in large part from animal studies, that the control of brown adipose tissue (BAT) thermogenesis is insured by the central nervous system (CNS), which integrates several stimuli in order to control BAT activation through the sympathetic nervous system (SNS). SNS-mediated BAT activity is governed by diverse neurons found in brain structures involved in homeostatic regulations and whose activity is modulated by various factors including oscillations of energy fluxes. The characterization of these neurons has always represented a challenging issue. The available literature suggests that the neuronal circuits controlling BAT thermogenesis are largely part of an autonomic circuitry involving the hypothalamus, brainstem and the SNS efferent neurons. In the present review, we recapitulate the latest progresses in regards to the hypothalamic regulation of BAT metabolism. We briefly addressed the role of the thermoregulatory pathway and its interactions with the energy balance systems in the control of thermogenesis. We also reviewed the involvement of the brain melanocortin and endocannabinoid systems as well as the emerging role of steroidogenic factor 1 (SF1) neurons in BAT thermogenesis. Finally, we examined the link existing between these systems and the homeostatic factors that modulate their activities.
Collapse
Affiliation(s)
- Sebastien M Labbé
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval Québec, QC, Canada
| | - Alexandre Caron
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval Québec, QC, Canada
| | - Damien Lanfray
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval Québec, QC, Canada
| | - Boris Monge-Rofarello
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval Québec, QC, Canada
| | - Timothy J Bartness
- Department of Biology, Center for Obesity Reversal (COR), Georgia State University Atlanta, GA, USA
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Department of Medicine, Université Laval Québec, QC, Canada
| |
Collapse
|
38
|
Bowden MA, Tesch GH, Julius TL, Rosli S, Love JE, Ritchie RH. Earlier onset of diabesity-Induced adverse cardiac remodeling in female compared to male mice. Obesity (Silver Spring) 2015; 23:1166-77. [PMID: 25959739 DOI: 10.1002/oby.21072] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/09/2015] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Emerging evidence suggests female type 2 diabetes (T2DM) patients may fare worse than males with respect to cardiovascular complications. Hence the impact of sex on relative progression of left ventricular (LV) remodeling in obese db/db mice was characterized. METHODS The changes in parameters of LV hypertrophy (heart weight, pro-hypertrophic gene expression, cardiomyocyte size) and fibrosis (LV collagen deposition and oxidative stress), in parallel with body weight and blood glucose and lipid profiles, in male and female db/db T2DM mice, at 10, 14, and 18 weeks of age, were determined. RESULTS Diabesity-induced cardiac remodeling was at least comparable in female (compared to male) mice. Females exhibited enhanced systemic oxidative stress and nonesterified fatty acid levels. Progression of LV pro-hypertrophic (β-myosin heavy chain, B-type natriuretic peptide) and pro-oxidant gene expression (NADPH oxidase subunit Nox2, plasminogen activator inhibitor-1 PAI-I) was, however, exaggerated in females when expressed relative to 10-week-old db/db mice. Increased cardiomyocyte width was also evident earlier in db/db females than males. No other gender differences were observed. CONCLUSIONS Progressive, age-dependent development of cardiac remodeling in db/db mice parallels impairments in glucose handling and oxidative stress. Certain aspects of the T2DM-induced LV remodeling response may have an earlier and/or exaggerated onset in diabetic females.
Collapse
Affiliation(s)
- Marissa A Bowden
- Heart Failure Pharmacology, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Greg H Tesch
- Department of Nephrology, Monash Medical Centre, Clayton, Victoria, Australia
- Department of Medicine, Monash University, Clayton, Victoria, Australia
| | - Tracey L Julius
- Heart Failure Pharmacology, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Sarah Rosli
- Heart Failure Pharmacology, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Jane E Love
- Heart Failure Pharmacology, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
- Department of Medicine, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
39
|
Tsuneki H, Tokai E, Nakamura Y, Takahashi K, Fujita M, Asaoka T, Kon K, Anzawa Y, Wada T, Takasaki I, Kimura K, Inoue H, Yanagisawa M, Sakurai T, Sasaoka T. Hypothalamic orexin prevents hepatic insulin resistance via daily bidirectional regulation of autonomic nervous system in mice. Diabetes 2015; 64:459-70. [PMID: 25249578 DOI: 10.2337/db14-0695] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Circadian rhythm is crucial for preventing hepatic insulin resistance, although the mechanism remains uncovered. Here we report that the wake-active hypothalamic orexin system plays a key role in this regulation. Wild-type mice showed that a daily rhythm in blood glucose levels peaked at the awake period; however, the glucose rhythm disappeared in orexin knockout mice despite normal feeding rhythm. Central administration of orexin A during nighttime awake period acutely elevated blood glucose levels but subsequently lowered daytime glucose levels in normal and diabetic db/db mice. The glucose-elevating and -lowering effects of orexin A were suppressed by adrenergic antagonists and hepatic parasympathectomy, respectively. Moreover, the expression levels of hepatic gluconeogenic genes, including Pepck, were increased and decreased by orexin A at nanomolar and femtomolar doses, respectively. These results indicate that orexin can bidirectionally regulate hepatic gluconeogenesis via control of autonomic balance, leading to generation of the daily blood glucose oscillation. Furthermore, during aging, orexin deficiency enhanced endoplasmic reticulum (ER) stress in the liver and caused impairment of hepatic insulin signaling and abnormal gluconeogenic activity in pyruvate tolerance test. Collectively, the daily glucose rhythm under control of orexin appears to be important for maintaining ER homeostasis, thereby preventing insulin resistance in the liver.
Collapse
Affiliation(s)
- Hiroshi Tsuneki
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Emi Tokai
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Yuya Nakamura
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Keisuke Takahashi
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Mikio Fujita
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Takehiro Asaoka
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Kanta Kon
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Yuuki Anzawa
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Tsutomu Wada
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| | - Ichiro Takasaki
- Division of Molecular Genetics Research, University of Toyama, Toyama, Japan
| | - Kumi Kimura
- Frontier Science Organization, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Hiroshi Inoue
- Frontier Science Organization, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan Howard Hughes Medical Institute, Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Takeshi Sakurai
- Department of Molecular Neuroscience and Integrative Physiology, Faculty of Medicine, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University of Toyama, Toyama, Japan
| |
Collapse
|
40
|
Katsuda Y, Sasase T, Tadaki H, Mera Y, Motohashi Y, Kemmochi Y, Toyoda K, Kakimoto K, Kume S, Ohta T. Contribution of hyperglycemia on diabetic complications in obese type 2 diabetic SDT fatty rats: effects of SGLT inhibitor phlorizin. Exp Anim 2015; 64:161-9. [PMID: 25736710 PMCID: PMC4427731 DOI: 10.1538/expanim.14-0084] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The spontaneously diabetic torii (SDT) fatty rat is a new model of type 2 diabetes
showing overt obesity, hyperglycemia and hyperlipidemia. With early onset of diabetes
mellitus, diabetic microvascular complications, including nephropathy, peripheral
neuropathy and retinopathy, are observed at young ages. In the present study, blood
glucose levels of female SDT fatty rats were controlled with phlorizin, a non-selective
SGLT inhibitor, to examine whether and how these complications are caused by
hyperglycemia. Phlorizin treatment adequately controlled plasma glucose levels during the
experiment. At 29 weeks of age, urinary albumin excretion considerably increased in SDT
fatty rats. Glomerulosclerosis and tubular pathological findings also indicate diabetic
nephropathy. These renal parameters tended to decrease with phlorizin; however, effects
were partial. Sciatic nerve conduction velocities were significantly delayed in SDT fatty
rats compared with Sprague-Dawley (SD) rats. Intraepidermal nerve fiber density, an
indicator of subclinical small nerve fiber neuropathy, significantly decreased in SDT
fatty rats. Retinal dysfunction (prolongation of peak latency for oscillatory potential in
electroretinograms) and histopathological eye abnormalities, including retinal folding and
mature cataracts were also observed. Both nerve and eye disorders were prevented with
phlorizin. These findings indicate that severe hyperglycemia mainly causes diabetic
complications in SDT fatty rats. However, other factors, such as hyperlipidemia and
hypertension, may affect diabetic nephropathy. These characteristics of diabetic
complications will become helpful in evaluating new drugs for diabetic complications using
SDT fatty rats.
Collapse
Affiliation(s)
- Yoshiaki Katsuda
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Soltysinska E, Speerschneider T, Winther SV, Thomsen MB. Sinoatrial node dysfunction induces cardiac arrhythmias in diabetic mice. Cardiovasc Diabetol 2014; 13:122. [PMID: 25113792 PMCID: PMC4149194 DOI: 10.1186/s12933-014-0122-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/03/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to probe cardiac complications, including heart-rate control, in a mouse model of type-2 diabetes. Heart-rate development in diabetic patients is not straight forward: In general, patients with diabetes have faster heart rates compared to non-diabetic individuals, yet diabetic patients are frequently found among patients treated for slow heart rates. Hence, we hypothesized that sinoatrial node (SAN) dysfunction could contribute to our understanding of the mechanism behind this conundrum and the consequences thereof. METHODS Cardiac hemodynamic and electrophysiological characteristics were investigated in diabetic db/db and control db/+ mice. RESULTS We found improved contractile function and impaired filling dynamics of the heart in db/db mice, relative to db/+ controls. Electrophysiologically, we observed comparable heart rates in the two mouse groups, but SAN recovery time was prolonged in diabetic mice. Adrenoreceptor stimulation increased heart rate in all mice and elicited cardiac arrhythmias in db/db mice only. The arrhythmias emanated from the SAN and were characterized by large RR fluctuations. Moreover, nerve density was reduced in the SAN region. CONCLUSIONS Enhanced systolic function and reduced diastolic function indicates early ventricular remodeling in obese and diabetic mice. They have SAN dysfunction, and adrenoreceptor stimulation triggers cardiac arrhythmia originating in the SAN. Thus, dysfunction of the intrinsic cardiac pacemaker and remodeling of the autonomic nervous system may conspire to increase cardiac mortality in diabetic patients.
Collapse
|
42
|
Abstract
Autonomic neuropathy complicates diabetes by increasing patient morbidity and mortality. Surprisingly, considering its importance, development and exploitation of animal models has lagged behind the wealth of information collected for somatic symmetrical sensory neuropathy. Nonetheless, animal studies have resulted in a variety of insights into the pathogenesis, neuropathology, and pathophysiology of diabetic autonomic neuropathy (DAN) with significant and, in some cases, remarkable correspondence between rodent models and human disease. Particularly in the study of alimentary dysfunction, findings in intrinsic intramural ganglia, interstitial cells of Cajal and the extrinsic parasympathetic and sympathetic ganglia serving the bowel vie for recognition as the chief mechanism. A body of work focused on neuropathologic findings in experimental animals and human subjects has demonstrated that axonal and dendritic pathology in sympathetic ganglia with relative neuron preservation represents one of the neuropathologic hallmarks of DAN but it is unlikely to represent the entire story. There is a surprising selectivity of the diabetic process for subpopulations of neurons and nerve terminals within intramural, parasympathetic, and sympathetic ganglia and innervation of end organs, afflicting some while sparing others, and differing between vascular and other targets within individual end organs. Rather than resulting from a simple deficit in one limb of an effector pathway, autonomic dysfunction may proceed from the inability to integrate portions of several complex pathways. The selectivity of the diabetic process appears to confound a simple global explanation (e.g., ischemia) of DAN. Although the search for a single unifying pathogenetic hypothesis continues, it is possible that autonomic neuropathy will have multiple pathogenetic mechanisms whose interplay may require therapies consisting of a cocktail of drugs. The role of multiple neurotrophic substances, antioxidants (general or pathway specific), inhibitors of formation of advanced glycosylation end products and drugs affecting the polyol pathway may be complex and therapeutic elements may have both salutary and untoward effects. This review has attempted to present the background and current findings and hypotheses, focusing on autonomic elements including and beyond the typical parasympathetic and sympathetic nervous systems to include visceral sensory and enteric nervous systems.
Collapse
Affiliation(s)
- Robert E Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
43
|
Verrotti A, Prezioso G, Scattoni R, Chiarelli F. Autonomic neuropathy in diabetes mellitus. Front Endocrinol (Lausanne) 2014; 5:205. [PMID: 25520703 PMCID: PMC4249492 DOI: 10.3389/fendo.2014.00205] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/17/2014] [Indexed: 12/14/2022] Open
Abstract
Diabetic autonomic neuropathy (DAN) is a serious and common complication of diabetes, often overlooked and misdiagnosed. It is a systemic-wide disorder that may be asymptomatic in the early stages. The most studied and clinically important form of DAN is cardiovascular autonomic neuropathy defined as the impairment of autonomic control of the cardiovascular system in patients with diabetes after exclusion of other causes. The reported prevalence of DAN varies widely depending on inconsistent definition, different diagnostic method, different patient cohorts studied. The pathogenesis is still unclear and probably multifactorial. Once DAN becomes clinically evident, no form of therapy has been identified, which can effectively stop or reverse it. Prevention strategies are based on strict glycemic control with intensive insulin treatment, multifactorial intervention, and lifestyle modification including control of hypertension, dyslipidemia, stop smoking, weight loss, and adequate physical exercise. The present review summarizes the latest knowledge regarding clinical presentation, epidemiology, pathogenesis, and management of DAN, with some mention to childhood and adolescent population.
Collapse
Affiliation(s)
| | | | | | - Francesco Chiarelli
- Department of Pediatrics, University of Chieti, Chieti, Italy
- *Correspondence: Francesco Chiarelli, Department of Pediatrics, University of Chieti, Via dei Vestini 5, Chieti 66013, Italy e-mail:
| |
Collapse
|
44
|
Abstract
Although obesity is a well-known risk factor for hypertension, the mechanisms by which hypertension develops in obese patients are not entirely clear. Animal models of obesity and their different susceptibilities to develop hypertension have revealed some of the mechanisms linking obesity and hypertension. Adipose tissue is an endocrine organ secreting hormones that impact blood pressure, such as elements of the renin-angiotensin system whose role in hypertension have been established. In addition, the appetite-suppressing adipokine leptin activates the sympathetic nervous system via the melanocortin system, and this activation, especially in the kidney, increases blood pressure. Leptin secretion from adipocytes is increased in most models of obesity due to leptin resistance, although the resistance is often selective to the anorexigenic effect, while the susceptibility to the hypertensive effect remains intact. Understanding the pathways by which obesity contributes to increased blood pressure will hopefully pave the way to and better define the appropriate treatment for obesity-induced hypertension.
Collapse
|
45
|
Ramos-Rodriguez JJ, Ortiz O, Jimenez-Palomares M, Kay KR, Berrocoso E, Murillo-Carretero MI, Perdomo G, Spires-Jones T, Cozar-Castellano I, Lechuga-Sancho AM, Garcia-Alloza M. Differential central pathology and cognitive impairment in pre-diabetic and diabetic mice. Psychoneuroendocrinology 2013; 38:2462-75. [PMID: 23790682 DOI: 10.1016/j.psyneuen.2013.05.010] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 04/11/2013] [Accepted: 05/13/2013] [Indexed: 12/12/2022]
Abstract
Although age remains the main risk factor to suffer Alzheimer's disease (AD) and vascular dementia (VD), type 2 diabetes (T2D) has turned up as a relevant risk factor for dementia. However, the ultimate underlying mechanisms for this association remain unclear. In the present study we analyzed central nervous system (CNS) morphological and functional consequences of long-term insulin resistance and T2D in db/db mice (leptin receptor KO mice). We also included C57Bl6 mice fed with high fat diet (HFD) and a third group of C57Bl6 streptozotocin (STZ) treated mice. Db/db mice exhibited pathological characteristics that mimic both AD and VD, including age dependent cognitive deterioration, brain atrophy, increased spontaneous hemorrhages and tau phosphorylation, affecting the cortex preferentially. A similar profile was observed in STZ-induced diabetic mice. Moreover metabolic parameters, such as body weight, glucose and insulin levels are good predictors of many of these alterations in db/db mice. In addition, in HFD-induced hyperinsulinemia in C57Bl6 mice, we only observed mild CNS alterations, suggesting that central nervous system dysfunction is associated with well established T2D. Altogether our results suggest that T2D may promote many of the pathological and behavioral alterations observed in dementia, supporting that interventions devoted to control glucose homeostasis could improve dementia progress and prognosis.
Collapse
|
46
|
Thackeray JT, deKemp RA, Beanlands RS, DaSilva JN. Insulin restores myocardial presynaptic sympathetic neuronal integrity in insulin-resistant diabetic rats. J Nucl Cardiol 2013; 20:845-56. [PMID: 23842711 DOI: 10.1007/s12350-013-9759-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 06/27/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Diabetes is associated with increased sympathetic activity, elevated norepinephrine, impaired heart rate variability, and the added risk of cardiovascular mortality. The temporal development of sympathetic neuronal dysfunction, response to therapy, and relation to ventricular function is not well characterized. METHODS AND RESULTS Sympathetic neuronal integrity was serially investigated in high fat diet-fed streptozotocin diabetic rats using [(11)C]meta-hydroxyephedrine (HED) positron emission tomography at baseline, 8 weeks of diabetes, and after a further 8 weeks of insulin or insulin-sensitizing metformin therapy. Myocardial HED retention was reduced in diabetic rats (n = 16) compared to non-diabetics (n = 6) at 8 weeks by 52-57% (P = .01) with elevated plasma and myocardial norepinephrine levels. Echocardiography pulse-wave Doppler measurements demonstrated prolonged mitral valve deceleration and increased early-to-atrial filling velocity, consistent with diastolic dysfunction. Insulin but not metformin evoked recovery of HED retention and plasma norepinephrine (P < .05), whereas echocardiography measurements of diastolic function were not improved by either treatment. Relative expressions of norepinephrine reuptake transporter and β-adrenoceptors were lower in metformin-treated as compared to insulin-treated diabetic and non-diabetic rats. Diabetic rats exhibited depressed heart rate variability and impaired diastolic function which persisted despite insulin treatment. CONCLUSIONS HED imaging provides sound estimation of sympathetic function. Effective glycemic control can recover sympathetic function in diabetic rats without the corresponding recovery of echocardiography indicators of diastolic dysfunction. HED positron emission tomography imaging may be useful in stratifying cardiovascular risk among diabetic patients and in evaluating the effect of glycemic therapy on the heart.
Collapse
Affiliation(s)
- James T Thackeray
- Molecular Function & Imaging Program, National Cardiac PET Centre, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON, K1Y 4W7, Canada,
| | | | | | | |
Collapse
|
47
|
Nakahara K, Bannai M, Maruyama K, Suzuki Y, Okame R, Murakami N. Characterization of a novel genetically obese mouse model demonstrating early onset hyperphagia and hyperleptinemia. Am J Physiol Endocrinol Metab 2013; 305:E451-63. [PMID: 23736543 DOI: 10.1152/ajpendo.00540.2012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Obesity is a critical risk factor for the development of metabolic syndrome, and many obese animal models are used to investigate the mechanisms responsible for the appearance of symptoms. To establish a new obese mouse model, we screened ∼13,000 ICR mice and discovered a mouse demonstrating spontaneous obesity. We named this mouse "Daruma" after a traditional Japanese ornament. Following the fixation of the genotype, these animals exhibited obese phenotypes according to Mendel's law of inheritance. In the Daruma mouse, the leptin receptor gene sequence carried two base mutations that are good candidates for the variation(s) responsible for the obese phenotype. The Daruma mice developed characteristic visceral fat accumulation at 4 wk of age, and the white adipose and liver tissues exhibited increases in cell size and lipid droplets, respectively. No histological abnormalities were observed in other tissues of the Daruma mice, even after the mice reached 25 wk of age. Moreover, the onset of impaired leptin signaling was early and manifested as hyperleptinemia and hyperinsulinemia. Pair feeding completely inhibited obesity, although these mice rapidly developed hyperphagia and obesity followed by hyperleptinemia when pair feeding ceased and free-access feeding was permitted. Therefore, the Daruma mice exhibited unique characteristics and may be a good model for studying human metabolic syndrome.
Collapse
Affiliation(s)
- Keiko Nakahara
- Department of Veterinary Physiology, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | | | | | | | | | | |
Collapse
|
48
|
Compensatory actions of orexinergic neurons in the lateral hypothalamus during metabolic or cortical challenges may enable the coupling of metabolic dysfunction and cortical dysfunction. Med Hypotheses 2013; 80:520-6. [DOI: 10.1016/j.mehy.2013.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 01/03/2013] [Accepted: 02/07/2013] [Indexed: 11/20/2022]
|
49
|
Sueta D, Kataoka K, Koibuchi N, Toyama K, Uekawa K, Katayama T, Mingjie M, Nakagawa T, Waki H, Maeda M, Yasuda O, Matsui K, Ogawa H, Kim-Mitsuyama S. Novel mechanism for disrupted circadian blood pressure rhythm in a rat model of metabolic syndrome--the critical role of angiotensin II. J Am Heart Assoc 2013; 2:e000035. [PMID: 23629805 PMCID: PMC3698757 DOI: 10.1161/jaha.113.000035] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND This study was performed to determine the characteristics and mechanism of hypertension in SHR/NDmcr-cp(+/+) rats (SHRcp), a new model of metabolic syndrome, with a focus on the autonomic nervous system, aldosterone, and angiotensin II. METHODS AND RESULTS We measured arterial blood pressure (BP) in SHRcp by radiotelemetry combined with spectral analysis using a fast Fourier transformation algorithm and examined the effect of azilsartan, an AT1 receptor blocker. Compared with control Wistar-Kyoto rats (WKY) and SHR, SHRcp exhibited a nondipper-type hypertension and displayed increased urinary norepinephrine excretion and increased urinary and plasma aldosterone levels. Compared with WKY and SHR, SHRcp were characterized by an increase in the low-frequency power (LF) of systolic BP and a decrease in spontaneous baroreflex gain (sBRG), indicating autonomic dysfunction. Thus, SHRcp are regarded as a useful model of human hypertension with metabolic syndrome. Oral administration of azilsartan once daily persistently lowered BP during the light period (inactive phase) and the dark period (active phase) in SHRcp more than in WKY and SHR. Thus, angiotensin II seems to be involved in the mechanism of disrupted diurnal BP rhythm in SHRcp. Azilsartan significantly reduced urinary norepinephrine and aldosterone excretion and significantly increased urinary sodium excretion in SHRcp. Furthermore, azilsartan significantly reduced LF of systolic BP and significantly increased sBRG in SHRcp. CONCLUSIONS These results strongly suggest that impairment of autonomic function and increased aldosterone in SHRcp mediate the effect of angiotensin II on circadian blood pressure rhythms.
Collapse
Affiliation(s)
- Daisuke Sueta
- Department of Pharmacology and Molecular Therapeutics, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Diabetic nephropathy is the single major cause of kidney failure in the industrialized world and given the emerging global pandemic of diabetes mellitus, its prevalence is expected to only increase. Because of the lack of dynamic biomarkers that define the rate of kidney function loss, there are few proof-of-concept clinical trials for new therapeutics to treat diabetic nephropathy. A molecular understanding of the pathogenesis of diabetic nephropathy also is lacking. These deficiencies are magnified by the fact that most mouse models of diabetic nephropathy fail to show progressive kidney disease. Recently, some mouse models that showed requisite phenotypic changes of diabetic nephropathy have been identified. Validation of results obtained in these experimental models, and showing whether they accurately can predict clinical response to therapeutics in human diabetic nephropathy, must now be established.
Collapse
Affiliation(s)
- Matthew D Breyer
- Lead Generation Biology, Biotechnology Discovery Research, Lilly Research Laboratories, Indianapolis, IN 46285, USA.
| |
Collapse
|