1
|
Lawton SB, Wagner VA, Nakagawa P, Segar JL, Sigmund CD, Morselli LL, Grobe JL. Angiotensin in the Arcuate: Mechanisms Integrating Cardiometabolic Control: The 2022 COH Mid-Career Award for Research Excellence. Hypertension 2024; 81:2209-2217. [PMID: 39315447 PMCID: PMC11483214 DOI: 10.1161/hypertensionaha.124.20524] [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] [Indexed: 09/25/2024]
Abstract
The American Heart Association has identified obesity as a primary impediment to ongoing improvements in cardiovascular diseases, including hypertension. Although drugs, exercise, diets, and surgeries can each cause weight loss, few subjects maintain a reduced weight over the long term. Dysfunctional integrative control (ie, adaptation) of resting metabolic rate (RMR) appears to underlie this failed weight maintenance, yet the neurobiology of physiological and pathophysiological RMR control is poorly understood. Here, we review recent insights into the cellular and molecular control of RMR by Ang-II (angiotensin II) signaling within the arcuate nucleus of the hypothalamus. Within a unique subset of agouti-related peptide neurons, AT1R (Ang-II type 1 receptors) are implicated in the integrative control of RMR. Furthermore, a spontaneous G protein signal switch of AT1R within this neuron type appears to underlie the pathogenesis of RMR adaptation by qualitatively changing the cellular response to AT1R activation from a β-arrestin-1/Gαi (heterotrimeric G protein, α i subtype)-mediated inhibitory response to a Gαq (heterotrimeric G protein, α q subtype)-mediated stimulatory response. We conclude that therapeutic approaches to obesity are likely hampered by the plasticity of the signaling mechanisms that mediate the normal integrative control of energy balance. The same stimulus that would increase RMR in the normal physiological state may decrease RMR during obesity due to qualitative changes in second-messenger coupling. Understanding the mechanisms that regulate interactions between receptors such as AT1R and its various second messenger signaling cascades will provide novel insights into the pathogenesis of RMR adaptation and potentially point toward new therapeutic approaches for obesity and hypertension.
Collapse
Affiliation(s)
- Samuel B.R. Lawton
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Valerie A. Wagner
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Jeffrey L. Segar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Lisa L. Morselli
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Medicine, Division of Endocrinology and Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
2
|
Ziegler AA, Lawton SBR, Grobe CC, Reho JJ, Freudinger BP, Burnett CML, Nakagawa P, Grobe JL, Segar JL. Early-life sodium deprivation programs long-term changes in ingestive behaviors and energy expenditure in C57BL/6J mice. Am J Physiol Regul Integr Comp Physiol 2023; 325:R576-R592. [PMID: 37720996 PMCID: PMC10866575 DOI: 10.1152/ajpregu.00137.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/19/2023]
Abstract
Postnatal growth failure remains a significant problem for infants born prematurely, despite aggressive efforts to improve perinatal nutrition. Though often dysregulated in early life when children are born preterm, sodium (Na) homeostasis is vital to achieve optimal growth. We hypothesize that insufficient Na supply in this critical period contributes to growth restriction and programmed risks for cardiometabolic disease in later adulthood. Thus, we sought to ascertain the effects of prolonged versus early-life Na depletion on weight gain, body composition, food and water intake behaviors, and energy expenditure in C57BL/6J mice. In one study, mice were provided a low (0.04%)- or normal/high (0.30%)-Na diet between 3 and 18 wk of age. Na-restricted mice demonstrated delayed growth and elevated basal metabolic rate. In a second study, mice were provided 0.04% or 0.30% Na diet between 3 and 6 wk of age and then returned to standard (0.15%)-Na diet through the end of the study. Na-restricted mice exhibited growth delays that quickly caught up on return to standard diet. Between 6 and 18 wk of age, previously restricted mice exhibited sustained, programmed changes in feeding behaviors, reductions in total food intake, and increases in water intake and aerobic energy expenditure while maintaining normal body composition. Although having no effect in control mice, administration of the ganglionic blocker hexamethonium abolished the programmed increase in basal metabolic rate in previously restricted mice. Together these data indicate that early-life Na restriction can cause programmed changes in ingestive behaviors, autonomic function, and energy expenditure that persist well into adulthood.
Collapse
Affiliation(s)
- Alisha A Ziegler
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Samuel B R Lawton
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Connie C Grobe
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Bonnie P Freudinger
- Engineering Core, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Colin M L Burnett
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Biomedical Engineering, Medical College of Wisconsin, Wisconsin, United States
| | - Jeffrey L Segar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Division of Neonatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
3
|
Mathieu NM, Fekete EM, Muskus PC, Brozoski DT, Lu KT, Wackman KK, Gomez J, Fang S, Reho JJ, Grobe CC, Vazirabad I, Mouradian GC, Hodges MR, Segar JL, Grobe JL, Sigmund CD, Nakagawa P. Genetic Ablation of Prorenin Receptor in the Rostral Ventrolateral Medulla Influences Blood Pressure and Hydromineral Balance in Deoxycorticosterone-Salt Hypertension. FUNCTION 2023; 4:zqad043. [PMID: 37609445 PMCID: PMC10440998 DOI: 10.1093/function/zqad043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Non-enzymatic activation of renin via its interaction with prorenin receptor (PRR) has been proposed as a key mechanism of local renin-angiotensin system (RAS) activation. The presence of renin and angiotensinogen has been reported in the rostral ventrolateral medulla (RVLM). Overactivation of bulbospinal neurons in the RVLM is linked to hypertension (HTN). Previous studies have shown that the brain RAS plays a role in the pathogenesis of the deoxycorticosterone (DOCA)-salt HTN model. Thus, we hypothesized that PRR in the RVLM is involved in the local activation of the RAS, facilitating the development of DOCA-salt HTN. Selective PRR ablation targeting the RVLM (PRRRVLM-Null mice) resulted in an unexpected sex-dependent and biphasic phenotype in DOCA-salt HTN. That is, PRRRVLM-Null females (but not males) exhibited a significant delay in achieving maximal pressor responses during the initial stage of DOCA-salt HTN. Female PRRRVLM-Null subsequently showed exacerbated DOCA-salt-induced pressor responses during the "maintenance" phase with a maximal peak at 13 d on DOCA-salt. This exacerbated response was associated with an increased sympathetic drive to the resistance arterioles and the kidney, exacerbated fluid and sodium intake and output in response to DOCA-salt, and induced mobilization of fluids from the intracellular to extracellular space concomitant with elevated vasopressin. Ablation of PRR suppressed genes involved in RAS activation and catecholamine synthesis in the RVLM but also induced expression of genes involved in inflammatory responses. This study illustrates complex and sex-dependent roles of PRR in the neural control of BP and hydromineral balance through autonomic and neuroendocrine systems. Graphical abstract.
Collapse
Affiliation(s)
- Natalia M Mathieu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Eva M Fekete
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Patricia C Muskus
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel T Brozoski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ko-Ting Lu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kelsey K Wackman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Javier Gomez
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shi Fang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John J Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Connie C Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ibrahim Vazirabad
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Gary C Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jeffrey L Segar
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Curt D Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
4
|
Pan S, Worker CJ, Feng Earley Y. The hypothalamus as a key regulator of glucose homeostasis: emerging roles of the brain renin-angiotensin system. Am J Physiol Cell Physiol 2023; 325:C141-C154. [PMID: 37273237 PMCID: PMC10312332 DOI: 10.1152/ajpcell.00533.2022] [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: 11/29/2022] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
The regulation of plasma glucose levels is a complex and multifactorial process involving a network of receptors and signaling pathways across numerous organs that act in concert to ensure homeostasis. However, much about the mechanisms and pathways by which the brain regulates glycemic homeostasis remains poorly understood. Understanding the precise mechanisms and circuits employed by the central nervous system to control glucose is critical to resolving the diabetes epidemic. The hypothalamus, a key integrative center within the central nervous system, has recently emerged as a critical site in the regulation of glucose homeostasis. Here, we review the current understanding of the role of the hypothalamus in regulating glucose homeostasis, with an emphasis on the paraventricular nucleus, the arcuate nucleus, the ventromedial hypothalamus, and lateral hypothalamus. In particular, we highlight the emerging role of the brain renin-angiotensin system in the hypothalamus in regulating energy expenditure and metabolic rate, as well as its potential importance in the regulation of glucose homeostasis.
Collapse
Affiliation(s)
- Shiyue Pan
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Caleb J Worker
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| | - Yumei Feng Earley
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Department of Physiology & Cell Biology, School of Medicine, University of Nevada, Reno, Reno, Nevada, United States
- Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada, United States
| |
Collapse
|
5
|
Brain Bioenergetics in Chronic Hypertension: Risk Factor for Acute Ischemic Stroke. Biochem Pharmacol 2022; 205:115260. [PMID: 36179931 DOI: 10.1016/j.bcp.2022.115260] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022]
Abstract
Chronic hypertension is one of the key modifiable risk factors for acute ischemic stroke, also contributing to determine greater neurological deficits and worse functional outcome when an acute cerebrovascular event would occur. A tight relationship exists between cerebrovascular autoregulation, neuronal activity and brain bioenergetics. In chronic hypertension, progressive adaptations of these processes occur as an attempt to cope with the demanding necessity of brain functions, creating a new steady-state homeostatic condition. However, these adaptive modifications are insufficient to grant an adequate response to possible pathological perturbations of the established fragile hemodynamic and metabolic homeostasis. In this narrative review, we will discuss the main mechanisms by which alterations in brain bioenergetics and mitochondrial function in chronic hypertension could lead to increased risk of acute ischemic stroke, stressing the interconnections between hemodynamic factors (i.e. cerebral autoregulation and neurovascular coupling) and metabolic processes. Both experimental and clinical pieces of evidence will be discussed. Moreover, the potential role of mitochondrial dysfunction in determining, or at least sustaining, the pathogenesis and progression of chronic neurogenic hypertension will be considered. In the perspective of novel therapeutic strategies aiming at improving brain bioenergetics, we propose some determinant factors to consider in future studies focused on the cause-effect relationships between chronic hypertension and brain bioenergetic abnormalities (and vice versa), so to help translational research in this so-far unfilled gap.
Collapse
|
6
|
Pioglitazone Synthetic Analogue Ameliorates Streptozotocin-Induced Diabetes Mellitus through Modulation of ACE 2/Angiotensin 1–7 via PI3K/AKT/mTOR Signaling Pathway. Pharmaceuticals (Basel) 2022; 15:ph15030341. [PMID: 35337139 PMCID: PMC8955304 DOI: 10.3390/ph15030341] [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: 02/11/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
The renin angiotensin aldosterone system has a localized key regulatory action, especially in liver and body circulation. Furthermore, it accomplishes a significant role in the downregulation of the PI3K/AKT/mTOR signaling pathway that is involved in type II diabetes mellitus pathogenesis. The current study aimed to evaluate the effect of a synthetic pioglitazone analogue (benzenesulfonamide derivative) compared to the standard pioglitazone hypoglycemic drug on enhancing liver insulin sensitivity via ACE 2/Ang (1–7)/PI3K/AKT/mTOR in experimental STZ-induced diabetes. After the model was established, rats were distributed into the normal control group, diabetic group, pioglitazone group (20 mg/kg), and a benzenesulfonamide derivative group (20 mg/kg), with the last 2 groups receiving oral treatment for 14 consecutive days. Our results suggested enhancing liver insulin sensitivity against the ACE2/Ang (1–7)/PI3K/AKT/mTOR pathway. Moreover, the synthetic compound produced a reduction in blood glucose levels, restored hyperinsulinemia back to normal, and enhanced liver glycogen deposition. In addition, it up regulated the ACE2/Ang (1–7)/PI3K/AKT/mTOR signaling pathway via increasing insulin receptor substrate 1 and 2 sensitivity to insulin, while it increased glucose transporter 2 expression in the rat pancreas. The study findings imply that the hypoglycemic effect of the benzenesulfonamide derivative is due to enhancing liver sensitivity to regulate blood glucose level via the ACE2/Ang (1–7)/PI3K/AKT/mTOR pathway.
Collapse
|
7
|
Hypothalamic Renin-Angiotensin System and Lipid Metabolism: Effects of Virgin Olive Oil versus Butter in the Diet. Nutrients 2021; 13:nu13020480. [PMID: 33572630 PMCID: PMC7912484 DOI: 10.3390/nu13020480] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/12/2022] Open
Abstract
The brain renin-angiotensin system (RAS) has been recently involved in the homeostatic regulation of energy. Our goal was to analyse the influence of a diet rich in saturated fatty acids (butter) against one enriched in monounsaturated fatty acids (olive oil) on hypothalamic RAS, and their relationship with the metabolism of fatty acids. Increases in body weight and visceral fat, together with an increase in aminopeptidase A expression and reductions in AngII and AngIV were observed in the hypothalamus of animals fed with the butter diet. In this group, a marked reduction in the expression of genes related to lipid metabolism (LPL, CD36, and CPT-1) was observed in liver and muscle. No changes were found in terms of body weight, total visceral fat and the expression of hepatic genes related to fatty acid metabolism in the olive oil diet. The expressions of LPL and CD36 were reduced in the muscles, although the decrease was lower than in the butter diet. At the same time, the fasting levels of leptin were reduced, no changes were observed in the hypothalamic expression of aminopeptidase A and decreases were noted in the levels of AngII, AngIV and AngIII. These results support that the type of dietary fat is able to modify the hypothalamic profile of RAS and the body energy balance, related to changes in lipid metabolism.
Collapse
|
8
|
Messiha BAS, Ali MRA, Khattab MM, Abo-Youssef AM. Perindopril ameliorates experimental Alzheimer's disease progression: role of amyloid β degradation, central estrogen receptor and hyperlipidemic-lipid raft signaling. Inflammopharmacology 2020; 28:1343-1364. [PMID: 32488543 DOI: 10.1007/s10787-020-00724-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/17/2020] [Indexed: 12/20/2022]
Abstract
Accumulating evidence indicates that over-stimulation of angiotensin-converting enzyme 1 (ACE1) activity is associated with β-amyloid (Aβ) and phosphorylated tau (p-tau)-induced apoptosis, oxido-nitrosative neuroinflammatory stress and neurodegeneration in Alzheimer's disease (AD). Alternatively, activation of the ACE2, the metalloprotease neprilysin (Neutral Endopeptidase; NEP) and the insulin-degrading enzyme (IDE) could oppose the effects of ACE1 activation. We aim to investigate the relationship between ACE1/ACE2/NEP/IDE and amyloidogenic/hyperlipidemic-lipid raft signaling in hyperlipidemic AD model. Induction of AD was performed in ovariectomized female rats with high-fat high fructose diet (HFFD) feeding after 4 weeks following D-galactose injection (150 mg/kg). The brain-penetrating ACE1 inhibitor perindopril (0.5 mg/kg/day, p.o.) was administered on a daily basis for 30 days. Perindopril significantly decreased hippocampal expression of ACE1 and increased expression of ACE2, NEP and IDE. Perindopril markedly decreased Aβ1-42, improved lipid profile and ameliorated the lipid raft protein markers caveolin1 (CAV1) and flotillin 1 (FLOT1). This was accompanied by decreased expression of p-tau and enhancement of cholinergic neurotransmission, coupled with decreased oxido-nitrosative neuroinflammatory stress, enhancement of blood-brain barrier (BBB) functioning and lower expression of the apoptotic markers glial fibrillary acidic protein (GFAP), Bax and β-tubulin. In addition, perindopril ameliorated histopathological damage and improved learning, cognitive and recognition impairment as well as depressive behavior in Morris water maze, Y maze, novel object recognition and forced swimming tests, respectively. Conclusively, perindopril could improve cognitive defects in AD rats, at least through activation of ACE2/NEP/IDE and inhibition of ACE1 and subsequent modulation of amyloidogenic/hyperlipidemic-lipid raft signaling and oxido-nitrosative stress.
Collapse
Affiliation(s)
- Basim A S Messiha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | - Mohammed R A Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| | - Amira M Abo-Youssef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
9
|
Sapouckey SA, Morselli LL, Deng G, Patil CN, Balapattabi K, Oliveira V, Claflin KE, Gomez J, Pearson NA, Potthoff MJ, Gibson-Corley KN, Sigmund CD, Grobe JL. Exploration of cardiometabolic and developmental significance of angiotensinogen expression by cells expressing the leptin receptor or agouti-related peptide. Am J Physiol Regul Integr Comp Physiol 2020; 318:R855-R869. [PMID: 32186897 DOI: 10.1152/ajpregu.00297.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Angiotensin II (ANG II) Agtr1a receptor (AT1A) is expressed in cells of the arcuate nucleus of the hypothalamus that express the leptin receptor (Lepr) and agouti-related peptide (Agrp). Agtr1a expression in these cells is required to stimulate resting energy expenditure in response to leptin and high-fat diets (HFDs), but the mechanism activating AT1A signaling by leptin remains unclear. To probe the role of local paracrine/autocrine ANG II generation and signaling in this mechanism, we bred mice harboring a conditional allele for angiotensinogen (Agt, encoding AGT) with mice expressing Cre-recombinase via the Lepr or Agrp promoters to cause cell-specific deletions of Agt (AgtLepr-KO and AgtAgrp-KO mice, respectively). AgtLepr-KO mice were phenotypically normal, arguing against a paracrine/autocrine AGT signaling mechanism for metabolic control. In contrast, AgtAgrp-KO mice exhibited reduced preweaning survival, and surviving adults exhibited altered renal structure and steroid flux, paralleling previous reports of animals with whole body Agt deficiency or Agt disruption in albumin (Alb)-expressing cells (thought to cause liver-specific disruption). Surprisingly, adult AgtAgrp-KO mice exhibited normal circulating AGT protein and hepatic Agt mRNA expression but reduced Agt mRNA expression in adrenal glands. Reanalysis of RNA-sequencing data sets describing transcriptomes of normal adrenal glands suggests that Agrp and Alb are both expressed in this tissue, and fluorescent reporter gene expression confirms Cre activity in adrenal gland of both Agrp-Cre and Alb-Cre mice. These findings lead to the iconoclastic conclusion that extrahepatic (i.e., adrenal) expression of Agt is critically required for normal renal development and survival.
Collapse
Affiliation(s)
- Sarah A Sapouckey
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Lisa L Morselli
- Division of Endocrinology, Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | - Guorui Deng
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Chetan N Patil
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | | | - Vanessa Oliveira
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Kristin E Claflin
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Javier Gomez
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nicole A Pearson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa.,Obesity Research & Education Initiative, University of Iowa, Iowa City, Iowa.,Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, Iowa
| | - Katherine N Gibson-Corley
- Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, Iowa.,Department of Pathology, University of Iowa, Iowa City, Iowa
| | - Curt D Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Justin L Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin.,Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
10
|
Benchoula K, Khatib A, Jaffar A, Ahmed QU, Sulaiman WMAW, Wahab RA, El-Seedi HR. The promise of zebrafish as a model of metabolic syndrome. Exp Anim 2019; 68:407-416. [PMID: 31118344 PMCID: PMC6842808 DOI: 10.1538/expanim.18-0168] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Metabolic syndrome is a cluster including hyperglycaemia, obesity, hypertension, and
hypertriglyceridaemia as a result of biochemical and physiological alterations and can
increase the risk of cardiovascular disease and diabetes. Fundamental research on this
disease requires validated animal models. One potential animal model that is rapidly
gaining in popularity is zebrafish (Danio rerio). The use of zebrafish as
an animal model conveys several advantages, including high human genetic homology,
transparent embryos and larvae that allow easier visualization. This review discusses how
zebrafish models contribute to the development of metabolic syndrome studies. Different
diseases in the cluster of metabolic syndrome, such as hyperglycaemia, obesity, diabetes,
and hypertriglyceridaemia, have been successfully studied using zebrafish; and the model
is promising for hypertension and cardiovascular metabolic-related diseases due to its
genetic similarity to mammals. Genetic mutation, chemical induction, and dietary
alteration are among the tools used to improve zebrafish models. This field is expanding,
and thus, more effective and efficient techniques are currently developed to fulfil the
increasing demand for thorough investigations.
Collapse
Affiliation(s)
- Khaled Benchoula
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Sultan Ahmad Shah Street, Kuantan 25200, Pahang, Malaysia
| | - Alfi Khatib
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Sultan Ahmad Shah Street, Kuantan 25200, Pahang, Malaysia.,Central Research and Animal Facility (CREAM), Kulliyyah of Science, International Islamic University Malaysia, Sultan Ahamad Shah Street, Kuantan 25200, Pahang, Malaysia
| | - Ashika Jaffar
- School of Biosciences & Technology, VIT University, Vellore 632014, India
| | - Qamar Udin Ahmed
- Pharmacognosy Research Group, Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Sultan Ahmad Shah Street, Kuantan 25200, Pahang, Malaysia
| | - Wan Mohd Azizi Wan Sulaiman
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Sultan Ahmad Shah Street, Kuantan 25200, Pahang, Malaysia
| | - Ridhwan Abd Wahab
- Kulliyah of Allied Health Science, International Islamic University Malaysia, Sultan Ahmad Shah Street, Kuantan 25200, Pahang, Malaysia
| | - Hesham R El-Seedi
- Pharmacognosy Group, Department of Medicinal Chemistry, Biomedical Centre, Uppsala University, Box 574, SE-751 23 Uppsala, Sweden.,Alrayan Medical colleges, Medina 42541, Kingdom of Saudi Arabia
| |
Collapse
|
11
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 663] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
12
|
El-Shoura EA, Messiha BA, Sharkawi SM, Hemeida RA. Perindopril ameliorates lipopolysaccharide-induced brain injury through modulation of angiotensin-II/angiotensin-1-7 and related signaling pathways. Eur J Pharmacol 2018; 834:305-317. [DOI: 10.1016/j.ejphar.2018.07.046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022]
|
13
|
Abdel-Fattah MM, Messiha BAS, Mansour AM. Modulation of brain ACE and ACE2 may be a promising protective strategy against cerebral ischemia/reperfusion injury: an experimental trial in rats. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1003-1020. [PMID: 29909460 DOI: 10.1007/s00210-018-1523-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022]
Abstract
The brain renin-angiotensin system (RAS) is considered a crucial regulator for physiological homeostasis and disease progression. We evaluated the protective effects of the angiotensin receptor blocker (ARB) telmisartan and the angiotensin-converting enzyme 2 (ACE2) activator xanthenone on experimental cerebral ischemia/reperfusion (I/R) injury. Rats were divided into a sham control, a cerebral I/R control, a standard treatment (nimodipine, 10 mg/kg/day, 15 days, p.o.), three telmisartan treatments (1, 3, and 10 mg/kg/day, 15 days, p.o.), and three xanthenone treatments (0.5, 1, and 2 mg/kg/day, 15 days, s.c.) groups. One hour after the last dose, all rats except the sham control group were exposed to 30-min cerebral ischemia followed by 24-h reperfusion. Brain ACE and ACE2 activities and the apoptotic marker caspase-3 levels were assessed. Glutathione (GSH), malondialdehyde (MDA), and nitric oxide end products (NOx) as oxidative markers and tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and IL-10 as immunological markers were assessed. Histopathological examination and immunohistochemical evaluation of glial fibrillary acidic protein (GFAP) were performed in cerebral cortex and hippocampus sections. Telmisartan and xanthenone in the higher doses restored MDA, NOx, TNF-α, IL-6, caspase-3, ACE, and GFAP back to normal levels and significantly increased GSH, IL-10, and ACE2 compared to I/R control values. Histopathologically, both agents showed mild degenerative changes and necrosis of neurons in cerebral cortex and hippocampus compared with I/R control group. Modulation of brain RAS, either through suppression of the classic ACE pathway or stimulation of its antagonist pathway ACE2, may be a promising strategy against cerebral I/R damage.
Collapse
Affiliation(s)
| | | | - Ahmed Mohamed Mansour
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
14
|
Morselli LL, Claflin KE, Cui H, Grobe JL. Control of Energy Expenditure by AgRP Neurons of the Arcuate Nucleus: Neurocircuitry, Signaling Pathways, and Angiotensin. Curr Hypertens Rep 2018; 20:25. [PMID: 29556733 DOI: 10.1007/s11906-018-0824-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Here, we review the current understanding of the functional neuroanatomy of neurons expressing Agouti-related peptide (AgRP) and the angiotensin 1A receptor (AT1A) within the arcuate nucleus (ARC) in the control of energy balance. RECENT FINDINGS The development and maintenance of obesity involves suppression of resting metabolic rate (RMR). RMR control is integrated via AgRP and proopiomelanocortin neurons within the ARC. Their projections to other hypothalamic and extrahypothalamic nuclei contribute to RMR control, though relatively little is known about the contributions of individual projections and the neurotransmitters involved. Recent studies highlight a role for AT1A, localized to AgRP neurons, but the specific function of AT1A within these cells remains unclear. AT1A functions within AgRP neurons to control RMR, but additional work is required to clarify its role within subpopulations of AgRP neurons projecting to distinct second-order nuclei, and the molecular mediators of its signaling within these cells.
Collapse
Affiliation(s)
- Lisa L Morselli
- Department of Pharmacology, University of Iowa, 51 Newton Rd., 2-307 BSB, Iowa City, IA, 52242, USA.,Department of Internal Medicine, Division of Endocrinology, University of Iowa, Iowa City, IA, 52242, USA
| | - Kristin E Claflin
- Department of Pharmacology, University of Iowa, 51 Newton Rd., 2-307 BSB, Iowa City, IA, 52242, USA
| | - Huxing Cui
- Department of Pharmacology, University of Iowa, 51 Newton Rd., 2-307 BSB, Iowa City, IA, 52242, USA.,Center for Hypertension Research, University of Iowa, Iowa City, IA, 52242, USA.,Obesity Research & Education Initiative, University of Iowa, Iowa City, IA, 52242, USA.,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52242, USA.,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA.,Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, 52242, USA
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa, 51 Newton Rd., 2-307 BSB, Iowa City, IA, 52242, USA. .,Center for Hypertension Research, University of Iowa, Iowa City, IA, 52242, USA. .,Obesity Research & Education Initiative, University of Iowa, Iowa City, IA, 52242, USA. .,Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52242, USA. .,Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA. .,Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW The influence of gut bacteria upon host physiology is increasingly recognized, but mechanistic links are lacking. Diseases of energetic imbalance such as obesity and diabetes represent major risk factors for cardiovascular diseases such as hypertension. Thus, here, we review current mechanistic contributions of the gut microbiota to host energetics. RECENT FINDINGS Gut bacteria generate a multitude of small molecules which can signal to host tissues within and beyond the gastrointestinal tract to influence host physiology, and gut bacteria can also influence host digestive efficiency by altering the bioavailability of polysaccharides, yet the quantitative energetic effects of these processes remain unclear. Recently, our team has demonstrated that gut bacteria constitute a major anaerobic thermogenic biomass, which can quantitatively account for obesity. Quantitative understanding of the mechanisms by which gut bacteria influence energy homeostasis may ultimately inform the relationship between gut bacteria and cardiovascular dysfunction.
Collapse
|
16
|
Sapouckey SA, Deng G, Sigmund CD, Grobe JL. Potential mechanisms of hypothalamic renin-angiotensin system activation by leptin and DOCA-salt for the control of resting metabolism. Physiol Genomics 2017; 49:722-732. [PMID: 28986397 DOI: 10.1152/physiolgenomics.00087.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/22/2017] [Indexed: 02/07/2023] Open
Abstract
The renin-angiotensin system (RAS), originally described as a circulating hormone system, is an enzymatic cascade in which the final vasoactive peptide angiotensin II (ANG) regulates cardiovascular, hydromineral, and metabolic functions. The RAS is also synthesized locally in a number of tissues including the brain, where it can act in a paracrine fashion to regulate blood pressure, thirst, fluid balance, and resting energy expenditure/resting metabolic rate (RMR). Recent studies demonstrate that ANG AT1A receptors (Agtr1a) specifically in agouti-related peptide (AgRP) neurons of the arcuate nucleus (ARC) coordinate autonomic and energy expenditure responses to various stimuli including deoxycorticosterone acetate (DOCA)-salt, high-fat feeding, and leptin. It remains unclear, however, how these disparate stimuli converge upon and activate this specific population of AT1A receptors in AgRP neurons. We hypothesize that these stimuli may act to stimulate local expression of the angiotensinogen (AGT) precursor for ANG, or the expression of AT1A receptors, and thereby local activity of the RAS within the (ARC). Here we review mechanisms that may control AGT and AT1A expression within the central nervous system, with a particular focus on mechanisms activated by steroids, dietary fat, and leptin.
Collapse
Affiliation(s)
- Sarah A Sapouckey
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Guorui Deng
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Curt D Sigmund
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Center for Hypertension Research, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Fraternal Order of Eagles' Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Justin L Grobe
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa; .,Molecular Medicine Graduate Program, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Center for Hypertension Research, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Fraternal Order of Eagles' Diabetes Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa.,Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa; and.,Obesity Research & Education Initiative, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
17
|
Khallaf WA, Messiha BA, Abo-Youssef AM, El-Sayed NS. Protective effects of telmisartan and tempol on lipopolysaccharide-induced cognitive impairment, neuroinflammation, and amyloidogenesis: possible role of brain-derived neurotrophic factor. Can J Physiol Pharmacol 2017; 95:850-860. [DOI: 10.1139/cjpp-2017-0042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Angiotensin II has pro-inflammatory and pro-oxidant potentials. We investigated the possible protective effects of the Angiotensin II receptor blocker telmisartan, compared with the superoxide scavenger tempol, on lipopolysaccharide (LPS)-induced cognitive decline and amyloidogenesis. Briefly, mice were allocated into a normal control group, an LPS control group, a tempol treatment group, and 2 telmisartan treatment groups. A behavioral study was conducted followed by a biochemical study via assessment of brain levels of beta amyloid (Aβ) and brain-derived neurotropic factor (BDNF) as amyloidogenesis and neuroplasticity markers, tumor necrosis factor alpha (TNF-α), nitric oxide end products (NOx), neuronal and inducible nitric oxide synthase (nNOS and iNOS) as inflammatory markers, and superoxide dismutase (SOD), malondialdehyde (MDA), glutathione reduced (GSH), and nitrotyrosine (NT) as oxido-nitrosative stress markers. Finally, histopathological examination of cerebral cortex, hippocampus, and cerebellum sections was performed using routine and special Congo red stains. Tempol and telmisartan improved cognition, decreased brain Aβ deposition and BDNF depletion, decreased TNF-α, NOx, nNOS, iNOS, MDA, and NT brain levels, and increased brain SOD and GSH contents, parallel to confirmatory histopathological evidences. In conclusion, tempol and telmisartan are promising drugs in managing cognitive impairment and amyloidogenesis, at least via upregulation of BDNF with inhibition of neuroinflammation and oxido-nitrosative stress.
Collapse
Affiliation(s)
- Waleed A.I. Khallaf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Basim A.S. Messiha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Amira M.H. Abo-Youssef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, Egypt
| | - Nesrine S. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| |
Collapse
|
18
|
Becker BK, Wang H, Zucker IH. Central TrkB blockade attenuates ICV angiotensin II-hypertension and sympathetic nerve activity in male Sprague-Dawley rats. Auton Neurosci 2017; 205:77-86. [PMID: 28549782 DOI: 10.1016/j.autneu.2017.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/17/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023]
Abstract
Increased sympathetic nerve activity and the activation of the central renin-angiotensin system are commonly associated with cardiovascular disease states such as hypertension and heart failure, yet the precise mechanisms contributing to the long-term maintenance of this sympatho-excitation are incompletely understood. Due to the established physiological role of neurotrophins contributing toward neuroplasticity and neuronal excitability along with recent evidence linking the renin-angiotensin system and brain-derived neurotrophic factor (BDNF) along with its receptor (TrkB), it is likely the two systems interact to promote sympatho-excitation during cardiovascular disease. However, this interaction has not yet been fully demonstrated, in vivo. Thus, we hypothesized that central angiotensin II (Ang II) treatment will evoke a sympatho-excitatory state mediated through the actions of BDNF/TrkB. We infused Ang II (20ng/min) into the right lateral ventricle of male Sprague-Dawley rats for twelve days with or without the TrkB receptor antagonist, ANA-12 (50ng/h). We found that ICV infusion of Ang II increased mean arterial pressure (+40.4mmHg), increased renal sympathetic nerve activity (+19.4% max activity), and induced baroreflex dysfunction relative to vehicle. Co-infusion of ANA-12 attenuated the increase in blood pressure (-20.6mmHg) and prevented the increase in renal sympathetic nerve activity (-22.2% max) and baroreflex dysfunction relative to Ang II alone. Ang II increased thirst and decreased food consumption, and Ang II+ANA-12 augmented the thirst response while attenuating the decrease in food consumption. We conclude that TrkB signaling is a mediator of the long-term blood pressure and sympathetic nerve activity responses to central Ang II activity. These findings demonstrate the involvement of neurotrophins such as BDNF in promoting Ang II-induced autonomic dysfunction and further implicate TrkB signaling in modulating presympathetic autonomic neurons during cardiovascular disease.
Collapse
Affiliation(s)
- Bryan K Becker
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Hanjun Wang
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE, USA.; Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Irving H Zucker
- Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA..
| |
Collapse
|
19
|
Theobald RJ. Role of centrally active cardiovascular agents in cognitive disorders. Curr Opin Pharmacol 2017; 33:70-75. [DOI: 10.1016/j.coph.2017.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/02/2017] [Accepted: 05/09/2017] [Indexed: 01/09/2023]
|
20
|
Claflin KE, Sandgren JA, Lambertz AM, Weidemann BJ, Littlejohn NK, Burnett CML, Pearson NA, Morgan DA, Gibson-Corley KN, Rahmouni K, Grobe JL. Angiotensin AT1A receptors on leptin receptor-expressing cells control resting metabolism. J Clin Invest 2017; 127:1414-1424. [PMID: 28263184 DOI: 10.1172/jci88641] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 01/12/2017] [Indexed: 12/13/2022] Open
Abstract
Leptin contributes to the control of resting metabolic rate (RMR) and blood pressure (BP) through its actions in the arcuate nucleus (ARC). The renin-angiotensin system (RAS) and angiotensin AT1 receptors within the brain are also involved in the control of RMR and BP, but whether this regulation overlaps with leptin's actions is unclear. Here, we have demonstrated the selective requirement of the AT1A receptor in leptin-mediated control of RMR. We observed that AT1A receptors colocalized with leptin receptors (LEPRs) in the ARC. Cellular coexpression of AT1A and LEPR was almost exclusive to the ARC and occurred primarily within neurons expressing agouti-related peptide (AgRP). Mice lacking the AT1A receptor specifically in LEPR-expressing cells failed to show an increase in RMR in response to a high-fat diet and deoxycorticosterone acetate-salt (DOCA-salt) treatments, but BP control remained intact. Accordingly, loss of RMR control was recapitulated in mice lacking AT1A in AgRP-expressing cells. We conclude that angiotensin activates divergent mechanisms to control BP and RMR and that the brain RAS functions as a major integrator for RMR control through its actions at leptin-sensitive AgRP cells of the ARC.
Collapse
|
21
|
Littlejohn NK, Keen HL, Weidemann BJ, Claflin KE, Tobin KV, Markan KR, Park S, Naber MC, Gourronc FA, Pearson NA, Liu X, Morgan DA, Klingelhutz AJ, Potthoff MJ, Rahmouni K, Sigmund CD, Grobe JL. Suppression of Resting Metabolism by the Angiotensin AT2 Receptor. Cell Rep 2016; 16:1548-1560. [PMID: 27477281 DOI: 10.1016/j.celrep.2016.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/09/2016] [Accepted: 07/01/2016] [Indexed: 11/15/2022] Open
Abstract
Activation of the brain renin-angiotensin system (RAS) stimulates energy expenditure through increasing of the resting metabolic rate (RMR), and this effect requires simultaneous suppression of the circulating and/or adipose RAS. To identify the mechanism by which the peripheral RAS opposes RMR control by the brain RAS, we examined mice with transgenic activation of the brain RAS (sRA mice). sRA mice exhibit increased RMR through increased energy flux in the inguinal adipose tissue, and this effect is attenuated by angiotensin II type 2 receptor (AT2) activation. AT2 activation in inguinal adipocytes opposes norepinephrine-induced uncoupling protein-1 (UCP1) production and aspects of cellular respiration, but not lipolysis. AT2 activation also opposes inguinal adipocyte function and differentiation responses to epidermal growth factor (EGF). These results highlight a major, multifaceted role for AT2 within inguinal adipocytes in the control of RMR. The AT2 receptor may therefore contribute to body fat distribution and adipose depot-specific effects upon cardio-metabolic health.
Collapse
Affiliation(s)
| | - Henry L Keen
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Kristin E Claflin
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin V Tobin
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Kathleen R Markan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Sungmi Park
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Meghan C Naber
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Nicole A Pearson
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Xuebo Liu
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA
| | - Curt D Sigmund
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA.
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
22
|
Ali MRAA, Abo-Youssef AMH, Messiha BAS, Khattab MM. Tempol and perindopril protect against lipopolysaccharide-induced cognition impairment and amyloidogenesis by modulating brain-derived neurotropic factor, neuroinflammation and oxido-nitrosative stress. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:637-56. [DOI: 10.1007/s00210-016-1234-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/21/2016] [Indexed: 01/01/2023]
|
23
|
Xue B, Yu Y, Zhang Z, Guo F, Beltz TG, Thunhorst RL, Felder RB, Johnson AK. Leptin Mediates High-Fat Diet Sensitization of Angiotensin II-Elicited Hypertension by Upregulating the Brain Renin-Angiotensin System and Inflammation. Hypertension 2016; 67:970-6. [PMID: 27021010 DOI: 10.1161/hypertensionaha.115.06736] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/04/2016] [Indexed: 12/21/2022]
Abstract
Obesity is characterized by increased circulating levels of the adipocyte-derived hormone leptin, which can increase sympathetic nerve activity and raise blood pressure. A previous study revealed that rats fed a high-fat diet (HFD) have an enhanced hypertensive response to subsequent angiotensin II administration that is mediated at least, in part, by increased activity of brain renin-angiotensin system and proinflammatory cytokines. This study tested whether leptin mediates this HFD-induced sensitization of angiotensin II-elicited hypertension by interacting with brain renin-angiotensin system and proinflammatory cytokine mechanisms. Rats fed an HFD for 3 weeks had significant increases in white adipose tissue mass, plasma leptin levels, and mRNA expression of leptin and its receptors in the lamina terminalis and hypothalamic paraventricular nucleus. Central infusion of a leptin receptor antagonist during HFD feeding abolished HFD sensitization of angiotensin II-elicited hypertension. Furthermore, central infusion of leptin mimicked the sensitizing action of HFD. Concomitant central infusions of the angiotensin II type 1 receptor antagonist irbesartan, the tumor necrosis factor-α synthesis inhibitor pentoxifylline, or the inhibitor of microglial activation minocycline prevented the sensitization produced by central infusion of leptin. RT-PCR analysis indicated that either HFD or leptin administration upregulated mRNA expression of several components of the renin-angiotensin system and proinflammatory cytokines in the lamina terminalis and paraventricular nucleus. The leptin antagonist and the inhibitors of angiotensin II type 1 receptor, tumor necrosis factor-α synthesis, and microglial activation all reversed the expression of these genes. The results suggest that HFD-induced sensitization of angiotensin II-elicited hypertension is mediated by leptin through upregulation of central renin-angiotensin system and proinflammatory cytokines.
Collapse
Affiliation(s)
- Baojian Xue
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.).
| | - Yang Yu
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.)
| | - Zhongming Zhang
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.)
| | - Fang Guo
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.)
| | - Terry G Beltz
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.)
| | - Robert L Thunhorst
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.)
| | - Robert B Felder
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.)
| | - Alan Kim Johnson
- From the Departments of Psychological and Brain Sciences (B.X., F.G., T.G.B., R.L.T., A.K.J.), Pharmacology (A.K.J.), and Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City; and Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Henan, China (Z.Z.).
| |
Collapse
|
24
|
Xue B, Thunhorst RL, Yu Y, Guo F, Beltz TG, Felder RB, Johnson AK. Central Renin-Angiotensin System Activation and Inflammation Induced by High-Fat Diet Sensitize Angiotensin II-Elicited Hypertension. Hypertension 2015; 67:163-70. [PMID: 26573717 DOI: 10.1161/hypertensionaha.115.06263] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/27/2015] [Indexed: 01/11/2023]
Abstract
Obesity has been shown to promote renin-angiotensin system activity and inflammation in the brain and to be accompanied by increased sympathetic activity and blood pressure. Our previous studies demonstrated that administration of a subpressor dose of angiotensin (Ang) II sensitizes subsequent Ang II-elicited hypertension. The present study tested whether high-fat diet (HFD) feeding also sensitizes the Ang II-elicited hypertensive response and whether HFD-induced sensitization is mediated by an increase in renin-angiotensin system activity and inflammatory mechanisms in the brain. HFD did not increase baseline blood pressure, but enhanced the hypertensive response to Ang II compared with a normal-fat diet. The sensitization produced by the HFD was abolished by concomitant central infusions of either a tumor necrosis factor-α synthesis inhibitor, pentoxifylline, an Ang II type 1 receptor blocker, irbesartan, or an inhibitor of microglial activation, minocycline. Furthermore, central pretreatment with tumor necrosis factor-α mimicked the sensitizing action of a central subpressor dose of Ang II, whereas central pentoxifylline or minocycline abolished this Ang II-induced sensitization. Real-time quantitative reverse transcription-polymerase chain reaction analysis of lamina terminalis tissue indicated that HFD feeding, central tumor necrosis factor-α, or a central subpressor dose of Ang II upregulated mRNA expression of several components of the renin-angiotensin system and proinflammatory cytokines, whereas inhibition of Ang II type 1 receptor and of inflammation reversed these changes. The results suggest that HFD-induced sensitization of Ang II-elicited hypertension is mediated by upregulation of the brain renin-angiotensin system and of central proinflammatory cytokines.
Collapse
Affiliation(s)
- Baojian Xue
- From the Departments of Psychological and Brain Sciences (B.X., R.L.T., F.G., T.G.B., A.K.J.), Pharmacology (A.K.J.), Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City.
| | - Robert L Thunhorst
- From the Departments of Psychological and Brain Sciences (B.X., R.L.T., F.G., T.G.B., A.K.J.), Pharmacology (A.K.J.), Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City
| | - Yang Yu
- From the Departments of Psychological and Brain Sciences (B.X., R.L.T., F.G., T.G.B., A.K.J.), Pharmacology (A.K.J.), Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City
| | - Fang Guo
- From the Departments of Psychological and Brain Sciences (B.X., R.L.T., F.G., T.G.B., A.K.J.), Pharmacology (A.K.J.), Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City
| | - Terry G Beltz
- From the Departments of Psychological and Brain Sciences (B.X., R.L.T., F.G., T.G.B., A.K.J.), Pharmacology (A.K.J.), Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City
| | - Robert B Felder
- From the Departments of Psychological and Brain Sciences (B.X., R.L.T., F.G., T.G.B., A.K.J.), Pharmacology (A.K.J.), Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City
| | - Alan Kim Johnson
- From the Departments of Psychological and Brain Sciences (B.X., R.L.T., F.G., T.G.B., A.K.J.), Pharmacology (A.K.J.), Internal Medicine (Y.Y., R.B.F.), and the François M. Abboud Cardiovascular Research Center (B.X., R.B.F., A.K.J.), University of Iowa, Iowa City
| |
Collapse
|
25
|
Littlejohn NK, Grobe JL. Opposing tissue-specific roles of angiotensin in the pathogenesis of obesity, and implications for obesity-related hypertension. Am J Physiol Regul Integr Comp Physiol 2015; 309:R1463-73. [PMID: 26491099 DOI: 10.1152/ajpregu.00224.2015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/15/2015] [Indexed: 12/24/2022]
Abstract
Metabolic disease, specifically obesity, has now become the greatest challenge to improving cardiovascular health. The renin-angiotensin system (RAS) exists as both a circulating hormone system and as a local paracrine signaling mechanism within various tissues including the brain, kidney, and adipose, and this system is strongly implicated in cardiovascular health and disease. Growing evidence also implicates the RAS in the control of energy balance, supporting the concept that the RAS may be mechanistically involved in the pathogenesis of obesity and obesity hypertension. Here, we review the involvement of the RAS in the entire spectrum of whole organism energy balance mechanisms, including behaviors (food ingestion and spontaneous physical activity) and biological processes (digestive efficiency and both aerobic and nonaerobic resting metabolic rates). We hypothesize that opposing, tissue-specific effects of the RAS to modulate these various components of energy balance can explain the apparently paradoxical results reported by energy-balance studies that involve stimulating, versus disrupting, the RAS. We propose a model in which such opposing and tissue-specific effects of the RAS can explain the failure of simple, global RAS blockade to result in weight loss in humans, and hypothesize that obesity-mediated uncoupling of endogenous metabolic rate control mechanisms can explain the phenomenon of obesity-related hypertension.
Collapse
Affiliation(s)
- Nicole K Littlejohn
- Department of Pharmacology, the Obesity Research and Education Initiative, the Fraternal Order of Eagles' Diabetes Research Center, the François M. Abboud Cardiovascular Research Center, and the Center for Hypertension Research, University of Iowa, Iowa City, Iowa
| | - Justin L Grobe
- Department of Pharmacology, the Obesity Research and Education Initiative, the Fraternal Order of Eagles' Diabetes Research Center, the François M. Abboud Cardiovascular Research Center, and the Center for Hypertension Research, University of Iowa, Iowa City, Iowa
| |
Collapse
|