1
|
Khan N, Kurnik-Łucka M, Latacz G, Gil K. Systematic-Narrative Hybrid Literature Review: Crosstalk between Gastrointestinal Renin-Angiotensin and Dopaminergic Systems in the Regulation of Intestinal Permeability by Tight Junctions. Int J Mol Sci 2024; 25:5566. [PMID: 38791603 PMCID: PMC11122119 DOI: 10.3390/ijms25105566] [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: 04/17/2024] [Revised: 05/12/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024] Open
Abstract
In the first part of this article, the role of intestinal epithelial tight junctions (TJs), together with gastrointestinal dopaminergic and renin-angiotensin systems, are narratively reviewed to provide sufficient background. In the second part, the current experimental data on the interplay between gastrointestinal (GI) dopaminergic and renin-angiotensin systems in the regulation of intestinal epithelial permeability are reviewed in a systematic manner using the PRISMA methodology. Experimental data confirmed the copresence of DOPA decarboxylase (DDC) and angiotensin converting enzyme 2 (ACE2) in human and rodent enterocytes. The intestinal barrier structure and integrity can be altered by angiotensin (1-7) and dopamine (DA). Both renin-angiotensin and dopaminergic systems influence intestinal Na+/K+-ATPase activity, thus maintaining electrolyte and nutritional homeostasis. The colocalization of B0AT1 and ACE2 indicates the direct role of the renin-angiotensin system in amino acid absorption. Yet, more studies are needed to thoroughly define the structural and functional interaction between TJ-associated proteins and GI renin-angiotensin and dopaminergic systems.
Collapse
Affiliation(s)
- Nadia Khan
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 31-008 Krakow, Poland
| | - Magdalena Kurnik-Łucka
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
| | - Gniewomir Latacz
- Faculty of Pharmacy, Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 31-008 Krakow, Poland
| | - Krzysztof Gil
- Faculty of Medicine, Department of Pathophysiology, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland
| |
Collapse
|
2
|
McArdle Z, Singh R, Bielefeldt-Ohmann H, Moritz K, Denton K, Schreuder M. Beneficial effects of brief early life angiotensin-converting enzyme inhibition wane with time in sheep with solitary functioning kidney. Clin Sci (Lond) 2023; 137:603-615. [PMID: 37018071 PMCID: PMC10116343 DOI: 10.1042/cs20220811] [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: 11/24/2022] [Revised: 03/26/2023] [Accepted: 04/05/2023] [Indexed: 04/23/2023]
Abstract
A child with a congenital solitary functioning kidney (SFK) may develop kidney disease from early in life due to hyperfiltration injury. Previously, we showed in a sheep model of SFK that brief angiotensin-converting enzyme inhibition (ACEi) early in life is reno-protective and increases renal functional reserve (RFR) at 8 months of age. Here we investigated the long-term effects of brief early ACEi in SFK sheep out to 20 months of age. At 100 days gestation (term = 150 days) SFK was induced by fetal unilateral nephrectomy, or sham surgery was performed (controls). SFK lambs received enalapril (SFK+ACEi; 0.5 mg/kg, once daily, orally) or vehicle (SFK) from 4 to 8 weeks of age. At 8, 14 and 20 months of age urinary albumin excretion was measured. At 20 months of age, we examined basal kidney function and RFR via infusion of combined amino acid and dopamine (AA+D). SFK+ACEi resulted in lower albuminuria (∼40%) at 8 months, but not at 14 or 20 months of age compared with vehicle-SFK. At 20 months, basal GFR (∼13%) was lower in SFK+ACEi compared with SFK, but renal blood flow (RBF), renal vascular resistance (RVR) and filtration fraction were similar to SFK. During AA+D, the increase in GFR was similar in SFK+ACEi and SFK animals, but the increase in RBF was greater (∼46%) in SFK+ACEi than SFK animals. Brief ACEi in SFK delayed kidney disease in the short-term but these effects were not sustained long-term.
Collapse
Affiliation(s)
- Zoe McArdle
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia
| | - Reetu R. Singh
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry & Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - Karen M. Moritz
- Child Health Research Centre and School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Kate M. Denton
- Cardiovascular Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia
- Correspondence: Kate M. Denton ()
| | - Michiel F. Schreuder
- Department of Pediatric Nephrology, Amalia Children’s Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
3
|
Interactions between the intrarenal dopaminergic and the renin-angiotensin systems in the control of systemic arterial pressure. Clin Sci (Lond) 2022; 136:1205-1227. [PMID: 35979889 DOI: 10.1042/cs20220338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/31/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022]
Abstract
Systemic arterial hypertension is one of the leading causes of morbidity and mortality in the general population, being a risk factor for many cardiovascular diseases. Although its pathogenesis is complex and still poorly understood, some systems appear to play major roles in its development. This review aims to update the current knowledge on the interaction of the intrarenal renin-angiotensin system (RAS) and dopaminergic system in the development of hypertension, focusing on recent scientific hallmarks in the field. The intrarenal RAS, composed of several peptides and receptors, has a critical role in the regulation of blood pressure (BP) and, consequently, the development of hypertension. The RAS is divided into two main intercommunicating axes: the classical axis, composed of angiotensin-converting enzyme, angiotensin II, and angiotensin type 1 receptor, and the ACE2/angiotensin-(1-7)/Mas axis, which appears to modulate the effects of the classical axis. Dopamine and its receptors are also increasingly showing an important role in the pathogenesis of hypertension, as abnormalities in the intrarenal dopaminergic system impair the regulation of renal sodium transport, regardless of the affected dopamine receptor subtype. There are five dopamine receptors, which are divided into two major subtypes: the D1-like (D1R and D5R) and D2-like (D2R, D3R, and D4R) receptors. Mice deficient in any of the five dopamine receptor subtypes have increased BP. Intrarenal RAS and the dopaminergic system have complex interactions. The balance between both systems is essential to regulate the BP homeostasis, as alterations in the control of both can lead to hypertension.
Collapse
|
4
|
Inoue K, Horwich T, Bhatnagar R, Bhatt K, Goldwater D, Seeman T, Watson KE. Urinary Stress Hormones, Hypertension, and Cardiovascular Events: The Multi-Ethnic Study of Atherosclerosis. Hypertension 2021; 78:1640-1647. [PMID: 34510914 DOI: 10.1161/hypertensionaha.121.17618] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kosuke Inoue
- Department of Epidemiology, UCLA Fielding School of Public Health (K.I., T.S.).,Department of Social Epidemiology, Graduate School of Medicine, Kyoto University, Japan (K.I.)
| | - Tamara Horwich
- Division of Cardiology (T.H., D.G., K.E.W.), Department of Medicine, University of California, Los Angeles
| | - Roshni Bhatnagar
- Department of Medicine, University of California, Los Angeles (R.B., K.B.)
| | - Karan Bhatt
- Department of Medicine, University of California, Los Angeles (R.B., K.B.)
| | - Deena Goldwater
- Division of Cardiology (T.H., D.G., K.E.W.), Department of Medicine, University of California, Los Angeles.,Division of Geriatrics (D.G., T.S.), Department of Medicine, University of California, Los Angeles
| | - Teresa Seeman
- Department of Epidemiology, UCLA Fielding School of Public Health (K.I., T.S.).,Division of Geriatrics (D.G., T.S.), Department of Medicine, University of California, Los Angeles
| | - Karol E Watson
- Division of Cardiology (T.H., D.G., K.E.W.), Department of Medicine, University of California, Los Angeles
| |
Collapse
|
5
|
Kobiec T, Otero-Losada M, Chevalier G, Udovin L, Bordet S, Menéndez-Maissonave C, Capani F, Pérez-Lloret S. The Renin-Angiotensin System Modulates Dopaminergic Neurotransmission: A New Player on the Scene. Front Synaptic Neurosci 2021; 13:638519. [PMID: 33967734 PMCID: PMC8100578 DOI: 10.3389/fnsyn.2021.638519] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/11/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is an extrapyramidal disorder characterized by neuronal degeneration in several regions of the peripheral and central nervous systems. It is the second most frequent neurodegenerative disease after Alzheimer's. It has become a major health problem, affecting 1% of the world population over 60 years old and 3% of people beyond 80 years. The main histological findings are intracellular Lewy bodies composed of misfolded α-synuclein protein aggregates and loss of dopaminergic neurons in the central nervous system. Neuroinflammation, apoptosis, mitochondrial dysfunction, altered calcium homeostasis, abnormal protein degradation, and synaptic pathobiology have been put forward as mechanisms leading to cell death, α-synuclein deposition, or both. A progressive loss of dopaminergic neurons in the substantia nigra late in the neurodegeneration leads to developing motor symptoms like bradykinesia, tremor, and rigidity. The renin-angiotensin system (RAS), which is involved in regulating blood pressure and body fluid balance, also plays other important functions in the brain. The RAS is involved in the autocrine and paracrine regulation of the nigrostriatal dopaminergic synapses. Dopamine depletion, as in PD, increases angiotensin II expression, which stimulates or inhibits dopamine synthesis and is released via AT1 or AT2 receptors. Furthermore, angiotensin II AT1 receptors inhibit D1 receptor activation allosterically. Therefore, the RAS may have an important modulating role in the flow of information from the brain cortex to the basal ganglia. High angiotensin II levels might even aggravate neurodegeneration, activating the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, which leads to increased reactive oxygen species production.
Collapse
Affiliation(s)
- Tamara Kobiec
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Guenson Chevalier
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Lucas Udovin
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Sofía Bordet
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Camila Menéndez-Maissonave
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía, Facultad de Psicología y Psicopedagogía, Universidad Católica Argentina, Buenos Aires, Argentina
- Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Biología, Universidad Argentina John F. Kennedy, Buenos Aires, Argentina
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago de Chile, Chile
| | - Santiago Pérez-Lloret
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Medicina, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
6
|
Campos J, Pacheco R. Involvement of dopaminergic signaling in the cross talk between the renin-angiotensin system and inflammation. Semin Immunopathol 2020; 42:681-696. [PMID: 32997225 PMCID: PMC7526080 DOI: 10.1007/s00281-020-00819-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) is a fundamental regulator of blood pressure and has emerged as an important player in the control of inflammatory processes. Accordingly, imbalance on RAS components either systemically or locally might trigger the development of inflammatory disorders by affecting immune cells. At the same time, alterations in the dopaminergic system have been consistently involved in the physiopathology of inflammatory disorders. Accordingly, the interaction between the RAS and the dopaminergic system has been studied in the context of inflammation of the central nervous system (CNS), kidney, and intestine, where they exert antagonistic actions in the regulation of the immune system. In this review, we summarized, integrated, and discussed the cross talk of the dopaminergic system and the RAS in the regulation of inflammatory pathologies, including neurodegenerative disorders, such as Parkinson’s disease. We analyzed the molecular mechanisms underlying the interaction between both systems in the CNS and in systemic pathologies. Moreover, we also analyzed the impact of the commensal microbiota in the regulation of RAS and dopaminergic system and how it is involved in inflammatory disorders. Furthermore, we summarized the therapeutic approaches that have yielded positive results in preclinical or clinical studies regarding the use of drugs targeting the RAS and dopaminergic system for the treatment of inflammatory conditions. Further understanding of the molecular and cellular regulation of the RAS-dopaminergic cross talk should allow the formulation of new therapies consisting of novel drugs and/or repurposing already existing drugs, alone or in combination, for the treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Javier Campos
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, 7780272 Ñuñoa, Santiago, Chile
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Fundación Ciencia & Vida, Av. Zañartu 1482, 7780272 Ñuñoa, Santiago, Chile. .,Universidad San Sebastián, 7510156 Providencia, Santiago, Chile.
| |
Collapse
|
7
|
Garcia-Garrote M, Perez-Villalba A, Garrido-Gil P, Belenguer G, Parga JA, Perez-Sanchez F, Labandeira-Garcia JL, Fariñas I, Rodriguez-Pallares J. Interaction between Angiotensin Type 1, Type 2, and Mas Receptors to Regulate Adult Neurogenesis in the Brain Ventricular-Subventricular Zone. Cells 2019; 8:E1551. [PMID: 31801296 PMCID: PMC6952803 DOI: 10.3390/cells8121551] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/21/2019] [Accepted: 11/28/2019] [Indexed: 12/30/2022] Open
Abstract
The renin-angiotensin system (RAS), and particularly its angiotensin type-2 receptors (AT2), have been classically involved in processes of cell proliferation and maturation during development. However, the potential role of RAS in adult neurogenesis in the ventricular-subventricular zone (V-SVZ) and its aging-related alterations have not been investigated. In the present study, we analyzed the role of major RAS receptors on neurogenesis in the V-SVZ of adult mice and rats. In mice, we showed that the increase in proliferation of cells in this neurogenic niche was induced by activation of AT2 receptors but depended partially on the AT2-dependent antagonism of AT1 receptor expression, which restricted proliferation. Furthermore, we observed a functional dependence of AT2 receptor actions on Mas receptors. In rats, where the levels of the AT1 relative to those of AT2 receptor are much lower, pharmacological inhibition of the AT1 receptor alone was sufficient in increasing AT2 receptor levels and proliferation in the V-SVZ. Our data revealed that interactions between RAS receptors play a major role in the regulation of V-SVZ neurogenesis, particularly in proliferation, generation of neuroblasts, and migration to the olfactory bulb, both in young and aged brains, and suggest potential beneficial effects of RAS modulators on neurogenesis.
Collapse
MESH Headings
- Age Factors
- Angiotensin II/metabolism
- Animals
- Immunohistochemistry
- Lateral Ventricles/metabolism
- Male
- Mice
- Mice, Knockout
- Models, Biological
- Neural Stem Cells/metabolism
- Neurogenesis/genetics
- Protein Binding
- Rats
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
Collapse
Affiliation(s)
- Maria Garcia-Garrote
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Ana Perez-Villalba
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Faculty of Psychology, Universidad Católica de Valencia, Valencia, 46100 Burjassot, Spain
| | - Pablo Garrido-Gil
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - German Belenguer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Juan A Parga
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Francisco Perez-Sanchez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Jose Luis Labandeira-Garcia
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Departamento de Biología Celular, Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Jannette Rodriguez-Pallares
- Laboratorio de Neurobiología Celular y Molecular de la Enfermedad de Parkinson, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Dpto. Ciencias Morfolóxicas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (M.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
8
|
Rukavina Mikusic NL, Kouyoumdzian NM, Uceda A, Del Mauro JS, Pandolfo M, Gironacci MM, Puyó AM, Toblli JE, Fernández BE, Choi MR. Losartan prevents the imbalance between renal dopaminergic and renin angiotensin systems induced by fructose overload. l-Dopa/dopamine index as new potential biomarker of renal dysfunction. Metabolism 2018; 85:271-285. [PMID: 29727629 DOI: 10.1016/j.metabol.2018.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/28/2018] [Accepted: 04/25/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND The renin angiotensin system (RAS) and the renal dopaminergic system (RDS) act as autocrine and paracrine systems to regulate renal sodium management and inflammation and their alterations have been associated to hypertension and renal damage. Nearly 30-50% of hypertensive patients have insulin resistance (IR), with a strong correlation between hyperinsulinemia and microalbuminuria. OBJECTIVE The aim of this study was to demonstrate the existence of an imbalance between RAS and RDS associated to IR, hypertension and kidney damage induced by fructose overload (FO), as well as to establish their prevention, by pharmacological inhibition of RAS with losartan. MATERIALS/METHODS Ninety-six male Sprague-Dawley rats were randomly divided into four groups and studied at 4, 8 and 12 weeks: control group (C4, C8 and C12; tap water to drink); fructose-overloaded group (F4, F8 and F12; 10% w/v fructose solution to drink); losartan-treated control (L) group (L4, L8 and L12; losartan 30 mg/kg/day, in drinking water); and fructose-overloaded plus losartan group (F + L4, F + L8 and F + L12, in fructose solution). RESULTS FO induced metabolic and hemodynamic alterations as well as an imbalance between RAS and RDS, characterized by increased renal angiotensin II levels and AT1R overexpression, reduced urinary excretion of dopamine, increased excretion of l-dopa (increased l-dopa/dopamine index) and down-regulation of D1R and tubular dopamine transporters OCT-2, OCT-N1 and total OCTNs. This imbalance was accompanied by an overexpression of renal tubular Na+, K+-ATPase, pro-inflammatory (NF-kB, TNF-α, IL-6) and pro-fibrotic (TGF-β1 and collagen) markers and by renal damage (microalbuminuria and reduced nephrin expression). Losartan prevented the metabolic and hemodynamic alterations induced by FO from week 4. Increased urinary l-dopa/dopamine index and decreased D1R renal expression associated to FO were also prevented by losartan since week 4. The same pattern was observed for renal expression of OCTs/OCTNs, Na+, K+-ATPase, pro-inflammatory and pro-fibrotic markers from week 8. The appearance of microalbuminuria and reduced nephrin expression was prevented by losartan at week 12. CONCLUSION The results of this study provide new insight regarding the mechanisms by which a pro-hypertensive and pro-inflammatory system, such as RAS, downregulates another anti-hypertensive and anti-inflammatory system such as RDS. Additionally, we propose the use of l-dopa/dopamine index as a biochemical marker of renal dysfunction in conditions characterized by sodium retention, IR and/or hypertension, and as a predictor of response to treatment and follow-up of these processes.
Collapse
Affiliation(s)
- Natalia Lucía Rukavina Mikusic
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina.
| | - Nicolás Martín Kouyoumdzian
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Ana Uceda
- Hospital Alemán, Laboratorio de Medicina Experimental, Av Pueyrredón 1640, C1118AAT CABA, Buenos Aires, Argentina
| | - Julieta Sofía Del Mauro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Farmacología, Cátedra de Farmacología, Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Marcela Pandolfo
- Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Mariela Mercedes Gironacci
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica, Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Ana María Puyó
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Jorge Eduardo Toblli
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Hospital Alemán, Laboratorio de Medicina Experimental, Av Pueyrredón 1640, C1118AAT CABA, Buenos Aires, Argentina
| | - Belisario Enrique Fernández
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina; Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Av. Gral Las Heras 2191, C1127AAD CABA, Buenos Aires, Argentina
| | - Marcelo Roberto Choi
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Junín 956, C1113AAD CABA, Buenos Aires, Argentina; Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Av. Gral Las Heras 2191, C1127AAD CABA, Buenos Aires, Argentina
| |
Collapse
|
9
|
Frame AA, Wainford RD. Mechanisms of altered renal sodium handling in age-related hypertension. Am J Physiol Renal Physiol 2018; 315:F1-F6. [PMID: 29442548 DOI: 10.1152/ajprenal.00594.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The prevalence of hypertension rises with age to approximately two out of three adults over the age of 60 in the United States. Although the mechanisms underlying age-related hypertension are incompletely understood, sodium homeostasis is critical to the long-term regulation of blood pressure and there is strong evidence that aging is associated with alterations in renal sodium handling. This minireview focuses on recent advancements in our understanding of the vascular, neurohumoral, and renal mechanisms that influence sodium homeostasis and promote age-related hypertension.
Collapse
Affiliation(s)
- Alissa A Frame
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine and The Whitaker Cardiovascular Institute , Boston, Massachusetts
| | - Richard D Wainford
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine and The Whitaker Cardiovascular Institute , Boston, Massachusetts
| |
Collapse
|
10
|
Garrido-Gil P, Rodriguez-Perez AI, Dominguez-Meijide A, Guerra MJ, Labandeira-Garcia JL. Bidirectional Neural Interaction Between Central Dopaminergic and Gut Lesions in Parkinson's Disease Models. Mol Neurobiol 2018; 55:7297-7316. [PMID: 29404956 DOI: 10.1007/s12035-018-0937-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 01/25/2018] [Indexed: 01/10/2023]
Abstract
The exact mechanism of gut dysfunction in Parkinson's disease and, conversely, the role of gut pathology in brain dopaminergic degeneration are controversial. We investigated the effects of nigral lesions on the colonic neurotransmission, the effect of gut inflammation on the nigrostriatal dopaminergic function, and the possible involvement of the vagus nerve and the local renin-angiotensin system (RAS). Nigrostriatal dopamine depletion was performed by bilateral injection 6-hydroxydopamine, and gut inflammation was induced by dextran sulfate sodium salt treatment in rats and mice, respectively, with or without vagal disruption. A decrease in central dopamine levels induced a decrease in colonic dopamine types 1 and 2 receptor expression together with an increase in the colonic levels of dopamine and a decrease in the levels of acetylcholine, which may explain a decrease in gut motility. Central dopaminergic depletion also induced an increase in the colonic levels of inflammatory and oxidative stress markers together with activation of the pro-inflammatory arm of the local RAS. Mice with acute (1 week) or subchronic (3 weeks) gut inflammation did not show a significant increase in colonic α-synuclein and phosphorylated α-synuclein expression during this relatively short survival period. Interestingly, we observed early changes in the nigrostriatal dopaminergic homeostasis, dopaminergic neuron death, and increased levels of nigral pro-inflammatory markers and RAS pro-inflammatory activity. The present results show that a dysregulation of the neural bidirectional gut-brain interaction may explain the early gut disturbances observed in parkinsonian patients, and also the increase in vulnerability of nigral dopaminergic neurons after gut inflammation.
Collapse
Affiliation(s)
- Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Antonio Dominguez-Meijide
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain. .,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
11
|
Garrido-Gil P, Dominguez-Meijide A, Moratalla R, Guerra MJ, Labandeira-Garcia JL. Aging-related dysregulation in enteric dopamine and angiotensin system interactions: implications for gastrointestinal dysfunction in the elderly. Oncotarget 2018. [PMID: 29541380 PMCID: PMC5834264 DOI: 10.18632/oncotarget.24330] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Gastrointestinal dysfunction is a common problem in the elderly. Aging-related changes in interactions between local dopaminergic and renin-angiotensin systems (RAS) have been observed in the brain, renal and vascular tissues. However, it is not known if these interactions also occur in the gut, and are dysregulated with aging. We showed a mutual regulation between the colonic dopaminergic system and RAS using young and aged mice deficient for major angiotensin and dopamine receptors. Aged rats showed a marked decrease in colonic dopamine D2 receptor expression, together with an increase in angiotensin type 1 (AT1) receptor expression, a decrease in angiotensin type 2 (AT2) receptor expression (i.e. an increase in the RAS pro-inflammatory arm activity), and increased levels of inflammatory and oxidative markers. Aged rats also showed increased levels of colonic dopamine and noradrenalin, and a marked decrease in acetylcholine and serotonin levels. The present observations contribute to explain an aging-related pro-inflammatory state and dysregulation in gastrointestinal function, which may be counteracted by treatment of aged animals with the AT1 receptor blocker candesartan.
Collapse
Affiliation(s)
- Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| | - Antonio Dominguez-Meijide
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| | - Rosario Moratalla
- Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain.,Instituto Cajal, Madrid, Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Research Center for Molecular Medicine and Chronic Diseases, University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Centre on Neurodegenerative Diseases, Madrid, Spain
| |
Collapse
|
12
|
Wu X, Fan R. Identifications of potential therapeutic targets and drugs in angiotensin II-induced hypertension. Medicine (Baltimore) 2017; 96:e8501. [PMID: 29145252 PMCID: PMC5704797 DOI: 10.1097/md.0000000000008501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/16/2022] Open
Abstract
This study aimed to identify the underlying therapeutic targets of angiotensin II (AngII)-induced hypertension, and screen the related drugs.The gene expression profiles of GSE93579 and GSE75815 were used to identify differentially expressed genes (DEGs) between AngII-induced hypertension and control samples based on meta-analysis. These DEGs were analyzed using Gene-Ontology (GO) function and pathway enrichment methods. Subsequently, the weighed gene coexpression network analysis (WGCNA)-based meta-analysis was applied to determine transcriptional signature with DEGs. Additionally, the functions of the modules were analyzed based on the network, and miRNAs were identified. Finally, small molecule drugs correlation with DEGs was identified.In total, 346 upregulated DEGs (e.g., Rgs7 bp) and 360 downregulated DEGs (e.g., Ebf3) were identified between AngII and control samples. In addition, a total of 150 DEGs in the brown, red, and yellow modules with higher correlation coefficient according to WGCNA, were used to construct the coexpression network, including Rgs7 bp and Ebf3, etc. in brown modules. Besides, 3 modules were obtained after the functions of the modules analysis. Moreover, 5 miRNAs were integrated in modules, including miR-124A, miR-524, miR-493, miR-323, and miR-203. Finally, anisomycin was the highest correlation with DEGs.MiR-124a might be involved in the pathogenesis of hypertension via targeting Ebf3 and Rgs7 bp, which possibly represent a novel and effective strategy for treatment of hypertension. Anisomycin might be performed to reduce blood pressure by blocking MAPK signaling pathway.
Collapse
Affiliation(s)
| | - Ruihua Fan
- Department of Medical Oncology, Huai’an First People's Hospital, Nanjing Medical University, Huai’an, Jiangsu, China
| |
Collapse
|
13
|
Dietary Fructose Enhances the Ability of Low Concentrations of Angiotensin II to Stimulate Proximal Tubule Na⁺ Reabsorption. Nutrients 2017; 9:nu9080885. [PMID: 28813008 PMCID: PMC5579678 DOI: 10.3390/nu9080885] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 11/18/2022] Open
Abstract
Fructose-enriched diets cause salt-sensitive hypertension. Proximal tubules (PTs) reabsorb 70% of the water and salt filtered through the glomerulus. Angiotensin II (Ang II) regulates this process. Normally, dietary salt reduces Ang II allowing the kidney to excrete more salt, thereby preventing hypertension. We hypothesized that fructose-enriched diets enhance the ability of low concentrations of Ang II to stimulate PT transport. We measured the effects of a low concentration of Ang II (10−12 mol/L) on transport-related oxygen consumption (QO2), and Na/K-ATPase and Na/H-exchange (NHE) activities and expression in PTs from rats consuming tap water (Control) or 20% fructose (FRUC). In FRUC-treated PTs, Ang II increased QO2 by 14.9 ± 1.3 nmol/mg/min (p < 0.01) but had no effect in Controls. FRUC elevated NHE3 expression by 19 ± 3% (p < 0.004) but not Na/K-ATPase expression. Ang II stimulated NHE activity in FRUC PT (Δ + 0.7 ± 0.1 Arbitrary Fluorescent units (AFU)/s, p < 0.01) but not in Controls. Na/K-ATPase activity was not affected. The PKC inhibitor Gö6976 blocked the ability of FRUC to augment the actions of Ang II. FRUC did not alter the inhibitory effect of dopamine on NHE activity. We conclude that dietary fructose increases the ability of low concentrations of Ang II to stimulate PT Na reabsorption via effects on NHE.
Collapse
|
14
|
Takahashi-Iwanaga H, Kimura S, Konno K, Watanabe M, Iwanaga T. Intrarenal signaling mediated by CCK plays a role in salt intake-induced natriuresis. Am J Physiol Renal Physiol 2017; 313:F20-F29. [DOI: 10.1152/ajprenal.00539.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 11/22/2022] Open
Abstract
The natriuretic hormone CCK exhibits its gene transcripts in total kidney extracts. To test the possibility of CCK acting as an intrarenal mediator of sodium excretion, we examined mouse kidneys by 1) an in situ hybridization technique for CCK mRNA in animals fed a normal- or a high-sodium diet; 2) immuno-electron microscopy for the CCK peptide, 3) an in situ hybridization method and immunohistochemistry for the CCK-specific receptor CCKAR; 4) confocal image analysis of receptor-mediated Ca2+ responses in isolated renal tubules; and 5) metabolic cage experiments for the measurement of urinary sodium excretion in high-salt-fed mice either treated or untreated with the CCKAR antagonist lorglumide. Results showed the CCK gene to be expressed intensely in the inner medulla and moderately in the inner stripe of the outer medulla, with the expression in the latter being enhanced by high sodium intake. Immunoreactivity for the CCK peptide was localized to the rough endoplasmic reticulum of the medullary interstitial cells in corresponding renal regions, confirming it to be a secretory protein. Gene transcripts, protein products, and the functional activity for CCKAR were consistently localized to the late proximal tubule segments (S2 and S3) in the medullary rays, and the outer stripe of the outer medulla. Lorglumide significantly diminished natriuretic responses of mice to a dietary sodium load without altering the glomerular filtration rate. These findings suggest that the medullary interstitial cells respond to body fluid expansion by CCK release for feedback regulation of the late proximal tubular reabsorption.
Collapse
Affiliation(s)
| | - Shunsuke Kimura
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kohtarou Konno
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshihiko Iwanaga
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
15
|
Dominguez-Meijide A, Rodriguez-Perez AI, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL. Dopamine modulates astroglial and microglial activity via glial renin-angiotensin system in cultures. Brain Behav Immun 2017; 62:277-290. [PMID: 28232171 DOI: 10.1016/j.bbi.2017.02.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 01/31/2017] [Accepted: 02/17/2017] [Indexed: 01/11/2023] Open
Abstract
Dopamine is an immunomodulatory molecule that acts on immune effector cells both in the CNS and peripheral tissues. However, the role of changes in dopamine levels in the neuroinflammatory response is controversial. The local/paracrine renin-angiotensin system (RAS) plays a major role in inflammatory processes in peripheral tissues and brain. In the present study, we investigated the possible role of the brain RAS in the effects of dopamine on the glial inflammatory responses. Astrocytes are the major source of the precursor protein angiotensinogen and angiotensin II (AII) in the brain. Neurotoxins such as MPP+ (1-methyl-4-phenylpyridinium) can act directly on astrocytes to increase levels of angiotensinogen and AII. Conversely, dopamine, via type-2 (D2) receptors, inhibited production of angiotensinogen, decreased expression of angiotensin type-1 (AT1) receptors and increased expression of AT2 receptors. In microglia, dopamine and dopamine agonists also regulated RAS activity. First, indirectly, via downregulation of the astrocyte-derived AII. Second, via dopamine-induced regulation of microglial angiotensin receptors. Dopamine decreased the microglial AT1/AT2 ratio leading to inhibition of the pro-inflammatory AT1/NADPH-oxidase/superoxide axis. D2 receptors were particularly responsible for microglial RAS inhibition in basal culture conditions. However, both D1 and D2 agonists inhibited the AT1/NADPH-oxidase axis in lipopolysaccharide-treated (LPS; i.e. activated) microglia. The results indicate that the decrease in dopamine levels observed in early stages of Parkinson's disease and aging may promote neuroinflammation and disease progression via glial RAS exacerbation.
Collapse
Affiliation(s)
- Antonio Dominguez-Meijide
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Carmen Diaz-Ruiz
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain.
| |
Collapse
|
16
|
Garrido-Gil P, Rodriguez-Perez AI, Fernandez-Rodriguez P, Lanciego JL, Labandeira-Garcia JL. Expression of angiotensinogen and receptors for angiotensin and prorenin in the rat and monkey striatal neurons and glial cells. Brain Struct Funct 2017; 222:2559-2571. [PMID: 28161727 DOI: 10.1007/s00429-016-1357-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/20/2016] [Indexed: 01/01/2023]
Abstract
The renin-angiotensin system (RAS) was initially considered as a circulating humoral system, which function is the regulation of blood pressure. However, it is now known that there exists local RAS in many tissues, including brain. In recent studies, we have demonstrated the presence of a local RAS in the substantia nigra of rodents and primates that modulates dopamine release and dopamine receptor expression. However, overactivation of local RAS exacerbates neuroinflammation, oxidative stress and dopaminergic cell death. In the striatum, it is not clear whether angiotensin receptors are located in dopaminergic terminals, glial cells and/or the projection neurons. The present study shows the location of major components of the RAS in striatal projection neurons of rats and monkeys (both in neurons of the direct and the indirect pathways). Striatal astrocytes and microglial cells also express major RAS components, which increase after induction of neuroinflammation by intrastriatal injection of lipopolysaccharide. Angiotensin receptors were located at the cell surface and also at cytoplasmic and nuclear levels. The results obtained by immunolabeling and confocal microscopy were confirmed with laser microdissection of striatal neurons and glial cells and detection of mRNA expression by PCR. The sequence of the resulting PCR products was verified by DNA sequencing. In addition to the interaction between angiotensin and dopamine receptors in dopaminergic neurons to regulate dopamine release, interaction between angiotensin and dopamine receptors in projection striatal neurons may further modulate the effects of dopamine on the direct and indirect pathways by fine-tuning striatal dopaminergic neurotransmission.
Collapse
Affiliation(s)
- Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Patricia Fernandez-Rodriguez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Lanciego
- Neurosciences Division, CIMA, University of Navarra, Pamplona, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
17
|
Ketchem CJ, Conner CD, Murray RD, DuPlessis M, Lederer ED, Wilkey D, Merchant M, Khundmiri SJ. Low dose ouabain stimulates NaK ATPase α1 subunit association with angiotensin II type 1 receptor in renal proximal tubule cells. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1863:2624-2636. [PMID: 27496272 PMCID: PMC5206903 DOI: 10.1016/j.bbamcr.2016.07.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/15/2016] [Accepted: 07/29/2016] [Indexed: 01/25/2023]
Abstract
Our laboratory has recently demonstrated that low concentrations of ouabain increase blood pressure in rats associated with stimulation of NaK ATPase activity and activation of the Src signaling cascade in NHE1-dependent manner. Proteomic analysis of human kidney proximal tubule cells (HKC11) suggested that the Angiotensin II type 1 receptor (AT1R) as an ouabain-associating protein. We hypothesize that ouabain-induced stimulation of NaK ATPase activity is mediated through AT1R. To test this hypothesis, we examined the effect of ouabain on renal cell angiotensin II production, the effect of AT1R inhibition on ouabain-stimulated NKA activity, and the effect of ouabain on NKA-AT1R association. Ouabain increased plasma angiotensin II levels in rats treated with ouabain (1μg/kg body wt./day) for 9days and increased angiotensin II levels in cell culture media after 24h treatment with ouabain in human (HKC11), mouse (MRPT), and human adrenal cells. Ouabain 10pM stimulated NKA-mediated 86Rb uptake and phosphorylation of EGFR, Src, and ERK1/2. These effects were prevented by the AT1R receptor blocker candesartan. FRET and TIRF microscopy using Bodipy-labeled ouabain and mCherry-NKA or mCherry-AT1R demonstrated association of ouabain with AT1R and NKA. Further our FRET and TIRF studies demonstrated increased association between AT1R and NKA upon treatment with low dose ouabain. We conclude that ouabain stimulates NKA in renal proximal tubule cells through an angiotensin/AT1R-dependent mechanism and that this pathway contributes to cardiac glycoside associated hypertension.
Collapse
Affiliation(s)
| | | | - Rebecca D Murray
- Department of Physiology, University of Louisville, KY, USA; Department of Medicine, University of Louisville, KY, USA
| | | | - Eleanor D Lederer
- Department of Physiology, University of Louisville, KY, USA; Department of Medicine, University of Louisville, KY, USA; Robley Rex VA Medical Center, Louisville, KY, USA
| | - Daniel Wilkey
- Department of Medicine, University of Louisville, KY, USA
| | | | - Syed J Khundmiri
- Department of Physiology and Biophysics, Howard University College of Medicine, Washington, DC, USA.
| |
Collapse
|
18
|
Labandeira-Garcia JL, Rodriguez-Perez AI, Valenzuela R, Costa-Besada MA, Guerra MJ. Menopause and Parkinson's disease. Interaction between estrogens and brain renin-angiotensin system in dopaminergic degeneration. Front Neuroendocrinol 2016; 43:44-59. [PMID: 27693730 DOI: 10.1016/j.yfrne.2016.09.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 02/07/2023]
Abstract
The neuroprotective effects of menopausal hormonal therapy in Parkinson's disease (PD) have not yet been clarified, and it is controversial whether there is a critical period for neuroprotection. Studies in animal models and clinical and epidemiological studies indicate that estrogens induce dopaminergic neuroprotection. Recent studies suggest that inhibition of the brain renin-angiotensin system (RAS) mediates the effects of estrogens in PD models. In the substantia nigra, ovariectomy induces a decrease in levels of estrogen receptor-α (ER-α) and increases angiotensin activity, NADPH-oxidase activity and expression of neuroinflammatory markers, which are regulated by estrogen replacement therapy. There is a critical period for the neuroprotective effect of estrogen replacement therapy, and local ER-α and RAS play a major role. Astrocytes play a major role in ER-α-induced regulation of local RAS, but neurons and microglia are also involved. Interestingly, treatment with angiotensin receptor antagonists after the critical period induced neuroprotection.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain.
| | - Ana I Rodriguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Rita Valenzuela
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Maria A Costa-Besada
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| |
Collapse
|
19
|
Burlaka I, Nilsson LM, Scott L, Holtbäck U, Eklöf AC, Fogo AB, Brismar H, Aperia A. Prevention of apoptosis averts glomerular tubular disconnection and podocyte loss in proteinuric kidney disease. Kidney Int 2016; 90:135-48. [PMID: 27217195 PMCID: PMC6101029 DOI: 10.1016/j.kint.2016.03.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 02/19/2016] [Accepted: 03/10/2016] [Indexed: 01/09/2023]
Abstract
There is a great need for treatment that arrests progression of chronic kidney disease. Increased albumin in urine leads to apoptosis and fibrosis of podocytes and tubular cells and is a major cause of functional deterioration. There have been many attempts to target fibrosis, but because of the lack of appropriate agents, few have targeted apoptosis. Our group has described an ouabain-activated Na,K-ATPase/IP3R signalosome, which protects from apoptosis. Here we show that albumin uptake in primary rat renal epithelial cells is accompanied by a time- and dose-dependent mitochondrial accumulation of the apoptotic factor Bax, down-regulation of the antiapoptotic factor Bcl-xL and mitochondrial membrane depolarization. Ouabain opposes these effects and protects from apoptosis in albumin-exposed proximal tubule cells and podocytes. The efficacy of ouabain as an antiapoptotic and kidney-protective therapeutic tool was then tested in rats with passive Heymann nephritis, a model of proteinuric chronic kidney disease. Chronic ouabain treatment preserved renal function, protected from renal cortical apoptosis, up-regulated Bax, down-regulated Bcl-xL, and rescued from glomerular tubular disconnection and podocyte loss. Thus we have identified a novel clinically feasible therapeutic tool, which has the potential to protect from apoptosis and rescue from loss of functional tissue in chronic proteinuric kidney disease.
Collapse
Affiliation(s)
- Ievgeniia Burlaka
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Linnéa M Nilsson
- Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Solna, Sweden
| | - Lena Scott
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
| | - Ulla Holtbäck
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Ann-Christine Eklöf
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Agnes B Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hjalmar Brismar
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden; Science for Life Laboratory, Department of Applied Physics, Royal Institute of Technology, Solna, Sweden
| | - Anita Aperia
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
20
|
Song J, Kim J. Degeneration of Dopaminergic Neurons Due to Metabolic Alterations and Parkinson's Disease. Front Aging Neurosci 2016; 8:65. [PMID: 27065205 PMCID: PMC4811934 DOI: 10.3389/fnagi.2016.00065] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/17/2016] [Indexed: 12/25/2022] Open
Abstract
The rates of metabolic diseases, such as type 2 diabetes mellitus (T2DM), obesity, and cardiovascular disease (CVD), markedly increase with age. In recent years, studies have reported an association between metabolic changes and various pathophysiological mechanisms in the central nervous system (CNS) in patients with metabolic diseases. Oxidative stress and hyperglycemia in metabolic diseases lead to adverse neurophysiological phenomena, including neuronal loss, synaptic dysfunction, and improper insulin signaling, resulting in Parkinson’s disease (PD). In addition, several lines of evidence suggest that alterations of CNS environments by metabolic changes influence the dopamine neuronal loss, eventually affecting the pathogenesis of PD. Thus, we reviewed recent findings relating to degeneration of dopaminergic neurons during metabolic diseases. We highlight the fact that using a metabolic approach to manipulate degeneration of dopaminergic neurons can serve as a therapeutic strategy to attenuate pathology of PD.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Biomedical Engineering, Dongguk University Seoul, South Korea
| | - Jongpil Kim
- Department of Biomedical Engineering, Dongguk University Seoul, South Korea
| |
Collapse
|
21
|
Wang Z, Zeng C, Villar VAM, Chen SY, Konkalmatt P, Wang X, Asico LD, Jones JE, Yang Y, Sanada H, Felder RA, Eisner GM, Weir MR, Armando I, Jose PA. Human GRK4γ142V Variant Promotes Angiotensin II Type I Receptor-Mediated Hypertension via Renal Histone Deacetylase Type 1 Inhibition. Hypertension 2015; 67:325-34. [PMID: 26667412 DOI: 10.1161/hypertensionaha.115.05962] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 11/03/2015] [Indexed: 12/14/2022]
Abstract
The influence of a single gene on the pathogenesis of essential hypertension may be difficult to ascertain, unless the gene interacts with other genes that are germane to blood pressure regulation. G-protein-coupled receptor kinase type 4 (GRK4) is one such gene. We have reported that the expression of its variant hGRK4γ(142V) in mice results in hypertension because of impaired dopamine D1 receptor. Signaling through dopamine D1 receptor and angiotensin II type I receptor (AT1R) reciprocally modulates renal sodium excretion and blood pressure. Here, we demonstrate the ability of the hGRK4γ(142V) to increase the expression and activity of the AT1R. We show that hGRK4γ(142V) phosphorylates histone deacetylase type 1 and promotes its nuclear export to the cytoplasm, resulting in increased AT1R expression and greater pressor response to angiotensin II. AT1R blockade and the deletion of the Agtr1a gene normalize the hypertension in hGRK4γ(142V) mice. These findings illustrate the unique role of GRK4 by targeting receptors with opposite physiological activity for the same goal of maintaining blood pressure homeostasis, and thus making the GRK4 a relevant therapeutic target to control blood pressure.
Collapse
Affiliation(s)
- Zheng Wang
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Chunyu Zeng
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Van Anthony M Villar
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Shi-You Chen
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Prasad Konkalmatt
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Xiaoyan Wang
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Laureano D Asico
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - John E Jones
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Yu Yang
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Hironobu Sanada
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Robin A Felder
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Gilbert M Eisner
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Matthew R Weir
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Ines Armando
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Pedro A Jose
- From the Division of Pediatric Nephrology, Department of Pediatrics, Georgetown University of School of Medicine, Washington, DC (Z.W.); Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China (C.Z.); Chongqing Institute of Cardiology, Chongqing, P.R. China; Division of Nephrology, Department of Medicine (V.A.M.V., X.W., L.D.A., J.E.J., Y.Y., M.R.W., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore, MD; Department of Physiology and Pharmacology, University of Georgia, Athens, GA (S.-Y.C.); Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan (H.S.); Department of Pathology, The University of Virginia Health Sciences Center, Charlottesville (R.A.F.); Department of Medicine, Georgetown University Medical Center, Washington, DC (G.M.E.); Division of Renal Diseases and Hypertension, Department of Medicine (P.A.J.) and Department of Physiology (P.A.J.), The George Washington University School of Medicine and Health Sciences, Washington, DC.
| |
Collapse
|
22
|
Giani JF, Bernstein KE, Janjulia T, Han J, Toblli JE, Shen XZ, Rodriguez-Iturbe B, McDonough AA, Gonzalez-Villalobos RA. Salt Sensitivity in Response to Renal Injury Requires Renal Angiotensin-Converting Enzyme. Hypertension 2015; 66:534-42. [PMID: 26150439 DOI: 10.1161/hypertensionaha.115.05320] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 06/15/2015] [Indexed: 12/24/2022]
Abstract
Recent evidence indicates that salt-sensitive hypertension can result from a subclinical injury that impairs the kidneys' capacity to properly respond to a high-salt diet. However, how this occurs is not well understood. Here, we showed that although previously salt-resistant wild-type mice became salt sensitive after the induction of renal injury with the nitric oxide synthase inhibitor Nω-nitro-l-arginine methyl ester hydrochloride; mice lacking renal angiotensin-converting enzyme, exposed to the same insult, did not become hypertensive when faced with a sodium load. This is because the activity of renal angiotensin-converting enzyme plays a critical role in (1) augmenting the local pool of angiotensin II and (2) the establishment of the antinatriuretic state via modulation of glomerular filtration rate and sodium tubular transport. Thus, this study demonstrates that the presence of renal angiotensin-converting enzyme plays a pivotal role in the development of salt sensitivity in response to renal injury.
Collapse
Affiliation(s)
- Jorge F Giani
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Kenneth E Bernstein
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Tea Janjulia
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Jiyang Han
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Jorge E Toblli
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Xiao Z Shen
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Bernardo Rodriguez-Iturbe
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Alicia A McDonough
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.)
| | - Romer A Gonzalez-Villalobos
- From the Departments of Biomedical Sciences and Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (J.F.G., K.E.B., T.J., X.Z.S., R.A.G.-V.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (J.H., A.A.M.); Laboratory of Experimental Medicine, Hospital Alemán, Buenos Aires, Argentina (J.E.T.); Servicio de Nefrología, Hospital Universitario de Maracaibo, Maracaibo, Venezuela (B.R.-I.); and Physiology Group, DSRD/Global Safety Pharmacology, Pfizer Inc., Groton, CT (R.A.G.-V.).
| |
Collapse
|
23
|
Kalra J, Prakash A, Kumar P, Majeed ABA. Cerebroprotective effects of RAS inhibitors: Beyond their cardio-renal actions. J Renin Angiotensin Aldosterone Syst 2015; 16:459-68. [PMID: 25944853 DOI: 10.1177/1470320315583582] [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: 04/02/2014] [Accepted: 03/22/2015] [Indexed: 01/25/2023] Open
Abstract
Work on the brain renin-angiotensin system has been explored by various researchers and has led to elucidation of its basic physiologies and behavior, including its role in reabsorption and uptake of body fluid, blood pressure maintenance with angiotensin II being its prominent effector. Currently, this system has been implicated for its newly established effects, which are far beyond its cardio-renal effects accounting for maintenance of cerebral blood flow and cerebroprotection, seizure, in the etiology of Alzheimer's disease, Parkinson's disease, multiple sclerosis, and bipolar disorder. In this review, we have discussed the distribution of angiotensin receptor subtypes in the central nervous system (CNS) together with enzymatic pathways leading to active angiotensin ligands and its interaction with angiotensin receptor 2 (AT2) and Mas receptors. Secondly, the use of angiotensin analogues (angiotensin converting enzyme inhibitors and AT1 and/or AT2 receptor blockers) in the treatment and management of the CNS disorders mentioned above has been discussed.
Collapse
Affiliation(s)
- Jaspreet Kalra
- Department of Pharmacology, Indo Soviet Friendship (ISF) College of Pharmacy, Moga, India
| | - Atish Prakash
- Department of Pharmacology, Indo Soviet Friendship (ISF) College of Pharmacy, Moga, India Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Malaysia Brain Degeneration and Therapeutics Group, Universiti Teknologi MARA (UiTM), Shah Alam, Malaysia
| | - Puneet Kumar
- Department of Pharmacology, Indo Soviet Friendship (ISF) College of Pharmacy, Moga, India
| | - Abu Bakar Abdul Majeed
- Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Malaysia Brain Degeneration and Therapeutics Group, Universiti Teknologi MARA (UiTM), Shah Alam, Malaysia
| |
Collapse
|
24
|
Activation of the D4 dopamine receptor attenuates proliferation and migration of vascular smooth muscle cells through downregulation of AT1a receptor expression. Hypertens Res 2015; 38:588-96. [PMID: 25832920 DOI: 10.1038/hr.2015.48] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 12/14/2014] [Accepted: 12/16/2014] [Indexed: 12/19/2022]
Abstract
Angiotensin (Ang) II has an important role in the vascular smooth muscle cell (VSMC) proliferation and migration and subsequently in the development of vascular diseases, whereas dopamine has the opposite effect. Previous studies have shown an interaction between dopamine and AT(1) receptors in the kidney. The dopamine D(4) receptor is expressed in arteries and has an inhibitory effect on VSMC proliferation. We hypothesized that the D(4) receptor, through its interaction with the AT(1a) receptor, may have an inhibitory effect on Ang II-mediated VSMC proliferation and migration, which could have a pivotal role in hypertension-induced vascular remodeling. In the current study, we found that Ang II markedly induced the proliferation and migration of A10 cells, which was inhibited by the D(4) receptor agonist PD168077. The activation of the D(4) receptor by PD168077 inhibited AT(1a) receptor expression in a concentration- and time-dependent manner. These effects were attenuated by silencing the D(4) receptor with a D(4) receptor-targeting small interfering RNA. The D(4) receptor-mediated inhibition of AT(1) receptor function involved protein kinase A (PKA). The activation of the D(4) receptor by PD168077 increased PKA activity in A10 cells, and the presence of a PKA inhibitor (PKA inhibitor 14-22, 10(-7) mol l(-1) per 24 h) blocked the inhibitory effect of the D(4) receptor on AT(1) receptor expression and function. The inhibitory effect of the D(4) receptor on AT(1) receptor expression and function was preserved in VSMCs (primary culture) from spontaneously hypertensive rats relative to VSMCs from Wistar-Kyoto rats. In conclusion, our data provide insight into the regulatory role of the D(4) receptor on AT(1a) receptor expression and function in VSMCs and suggest that targeting the action of the D(4) receptor may represent an effective therapeutic approach for the treatment of cardiovascular diseases.
Collapse
|
25
|
Signaling pathways involved in renal oxidative injury: role of the vasoactive peptides and the renal dopaminergic system. JOURNAL OF SIGNAL TRANSDUCTION 2014; 2014:731350. [PMID: 25436148 PMCID: PMC4243602 DOI: 10.1155/2014/731350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/16/2014] [Indexed: 12/24/2022]
Abstract
The physiological hydroelectrolytic balance and the redox steady state in the kidney are accomplished by an intricate interaction between signals from extrarenal and intrarenal sources and between antinatriuretic and natriuretic factors. Angiotensin II, atrial natriuretic peptide and intrarenal dopamine play a pivotal role in this interactive network. The balance between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide, by one side, and the prooxidant effect of the renin angiotensin system, by the other side, contributes to ensuring the normal function of the kidney. Different pathological scenarios, as nephrotic syndrome and hypertension, where renal sodium excretion is altered, are associated with an impaired interaction between two natriuretic systems as the renal dopaminergic system and atrial natriuretic peptide that may be involved in the pathogenesis of renal diseases. The aim of this review is to update and comment the most recent evidences about the intracellular pathways involved in the relationship between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide and the prooxidant effect of the renin angiotensin system in the pathogenesis of renal inflammation.
Collapse
|
26
|
Muñoz A, Garrido-Gil P, Dominguez-Meijide A, Labandeira-Garcia JL. Angiotensin type 1 receptor blockage reduces l-dopa-induced dyskinesia in the 6-OHDA model of Parkinson's disease. Involvement of vascular endothelial growth factor and interleukin-1β. Exp Neurol 2014; 261:720-32. [DOI: 10.1016/j.expneurol.2014.08.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 08/01/2014] [Accepted: 08/16/2014] [Indexed: 12/17/2022]
|
27
|
Ennis RC, Asico LD, Armando I, Yang J, Feranil JB, Jurgens JA, Escano CS, Yu P, Wang X, Sibley DR, Jose PA, Villar VAM. Dopamine D₁-like receptors regulate the α₁A-adrenergic receptor in human renal proximal tubule cells and D₁-like dopamine receptor knockout mice. Am J Physiol Renal Physiol 2014; 307:F1238-48. [PMID: 25339698 DOI: 10.1152/ajprenal.00119.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The homeostatic control of blood pressure hinges upon the delicate balance between prohypertensinogenic and antihypertensinogenic systems. D₁-like dopamine receptors [dopamine D₁ and D₅ receptors (D₁Rs and D₅Rs, respectively)] and the α₁A-adrenergic receptor (α₁A-AR) are expressed in the renal proximal tubule and engender opposing effects on Na(+) transport, i.e., natriuresis (via D₁Rs and D5Rs) or antinatriuresis (via α₁A-ARs). We tested the hypothesis that the D₁R/D₅R regulates the α₁A-AR. D₁-like dopamine receptors coimmunoprecipitated, colocalized, and cofractionated with α₁A-ARs in lipid rafts in immortalized human renal proximal tubule cells. Long-term treatment with the D₁R/D₅R agonist fenoldopam resulted in decreased D₁R and D₅R expression but increased α₁A-AR abundance in the plasma membrane. Short-term fenoldopam treatment stimulated the translocation of Na(+)-K(+)-ATPase from the plasma membrane to the cytosol that was partially reversed by an α₁A-AR agonist, which by itself induced Na(+)-K(+)-ATPase translocation from the cytosol to the plasma membrane. The α₁A-AR-specific agonist A610603 also minimized the ability of fenoldopam to inhibit Na(+)-K(+)-ATPase activity. To determine the interaction among D₁Rs, D₅Rs, and α₁A-ARs in vivo, we used phenylephrine and A610603 to decrease Na(+) excretion in several D1-like dopamine receptor knockout mouse strains. Phenylephrine and A61603 treatment resulted in a partial reduction of urinary Na(+) excretion in wild-type mice and its abolition in D1R knockout, D₅R knockout, and D₁R-D₅R double-knockout mice. Our results demonstrate the ability of the D₁-like dopamine receptors to regulate the expression and activity of α₁A-AR. Elucidating the intricacies of the interaction among these receptors is crucial for a better understanding of the crosstalk between anti- and pro-hypertensive systems.
Collapse
Affiliation(s)
- Riley Charles Ennis
- Thomas Jefferson High School for Science and Technology, Alexandria, Virgina
| | - Laureano D Asico
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jian Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jun B Feranil
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Julie A Jurgens
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Crisanto S Escano
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peiying Yu
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Xiaoyan Wang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Van Anthony M Villar
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland;
| |
Collapse
|
28
|
Borrajo A, Rodriguez-Perez AI, Diaz-Ruiz C, Guerra MJ, Labandeira-Garcia JL. Microglial TNF-α mediates enhancement of dopaminergic degeneration by brain angiotensin. Glia 2014; 62:145-57. [PMID: 24272709 DOI: 10.1002/glia.22595] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 10/14/2013] [Accepted: 10/16/2013] [Indexed: 11/11/2022]
Abstract
In vitro and in vivo models of Parkinson's disease were used to investigate whether TNF-α plays a major role in the enhancement of the microglial response and dopaminergic degeneration induced by brain angiotensin hyperactivity. Treatment of primary mesencephalic cultures with low doses of the neurotoxin MPP(+) induced a significant loss of dopaminergic neurons, which was enhanced by cotreatment with angiotensin II and inhibited by TNF-α inhibitors. Treatment of primary cultures with angiotensin induced a marked increase in levels of TNF-α, which was inhibited by treatment with angiotensin type-1-receptor antagonists, NADPH-oxidase inhibitors and NFK-β inhibitors. However, TNF-α levels were not significantly affected by treatment with angiotensin in the absence of microglia. The microglial origin of the angiotensin-induced increase in TNF-α levels was confirmed using dopaminergic (MES 23.5) and microglial (N9) cell lines. Inhibition of the microglial Rho-kinase activity also blocked the AII-induced increase in TNF-α levels. Treatment of the dopaminergic cell line with TNF-α revealed that NFK-β activation mediates the deleterious effect of microglial TNF-α on dopaminergic neurons. Treatment of mice with MPTP also induced significant increases in striatal and nigral TNF-α levels, which were inhibited by angiotensin type-1-receptor antagonists or NFK-β inhibitors. The present results show that microglial TNF-α plays a major role in angiotensin-induced dopaminergic cell death and that the microglial release of TNF-α is mediated by activation of angiotensin type-1 receptors, NADPH-oxidase, Rho-kinase and NFK-β.
Collapse
Affiliation(s)
- Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, Santiago de Compostela, Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | | | | | | | | |
Collapse
|
29
|
Labandeira-García JL, Garrido-Gil P, Rodriguez-Pallares J, Valenzuela R, Borrajo A, Rodríguez-Perez AI. Brain renin-angiotensin system and dopaminergic cell vulnerability. Front Neuroanat 2014; 8:67. [PMID: 25071471 PMCID: PMC4086395 DOI: 10.3389/fnana.2014.00067] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 06/24/2014] [Indexed: 01/11/2023] Open
Abstract
Although the renin-angiotensin system (RAS) was classically considered as a circulating system that regulates blood pressure, many tissues are now known to have a local RAS. Angiotensin, via type 1 receptors, is a major activator of the NADPH-oxidase complex, which mediates several key events in oxidative stress (OS) and inflammatory processes involved in the pathogenesis of major aging-related diseases. Several studies have demonstrated the presence of RAS components in the basal ganglia, and particularly in the nigrostriatal system. In the nigrostriatal system, RAS hyperactivation, via NADPH-oxidase complex activation, exacerbates OS and the microglial inflammatory response and contributes to progression of dopaminergic degeneration, which is inhibited by angiotensin receptor blockers and angiotensin converting enzyme (ACE) inhibitors. Several factors may induce an increase in RAS activity in the dopaminergic system. A decrease in dopaminergic activity induces compensatory upregulation of local RAS function in both dopaminergic neurons and glia. In addition to its role as an essential neurotransmitter, dopamine may also modulate microglial inflammatory responses and neuronal OS via RAS. Important counterregulatory interactions between angiotensin and dopamine have also been observed in several peripheral tissues. Neurotoxins and proinflammatory factors may also act on astrocytes to induce an increase in RAS activity, either independently of or before the loss of dopamine. Consistent with a major role of RAS in dopaminergic vulnerability, increased RAS activity has been observed in the nigra of animal models of aging, menopause and chronic cerebral hypoperfusion, which also showed higher dopaminergic vulnerability. Manipulation of the brain RAS may constitute an effective neuroprotective strategy against dopaminergic vulnerability and progression of Parkinson's disease.
Collapse
Affiliation(s)
- Jose L Labandeira-García
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Jannette Rodriguez-Pallares
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Rita Valenzuela
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Ana Borrajo
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| | - Ana I Rodríguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, CIMUS, Faculty of Medicine, University of Santiago de Compostela Santiago de Compostela, Spain ; Networking Research Center on Neurodegenerative Diseases (CIBERNED) Madrid, Spain
| |
Collapse
|
30
|
Aging-related dysregulation of dopamine and angiotensin receptor interaction. Neurobiol Aging 2014; 35:1726-38. [PMID: 24529758 DOI: 10.1016/j.neurobiolaging.2014.01.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 01/10/2014] [Accepted: 01/13/2014] [Indexed: 12/22/2022]
Abstract
It is not known whether the aging-related decrease in dopaminergic function leads to the aging-related higher vulnerability of dopaminergic neurons and risk for Parkinson's disease. The renin-angiotensin system (RAS) plays a major role in the inflammatory response, neuronal oxidative stress, and dopaminergic vulnerability via type 1 (AT1) receptors. In the present study, we observed a counterregulatory interaction between dopamine and angiotensin receptors. We observed overexpression of AT1 receptors in the striatum and substantia nigra of young adult dopamine D1 and D2 receptor-deficient mice and young dopamine-depleted rats, together with compensatory overexpression of AT2 receptors or compensatory downregulation of angiotensinogen and/or angiotensin. In aged rats, we observed downregulation of dopamine and dopamine receptors and overexpression of AT1 receptors in aged rats, without compensatory changes observed in young animals. L-Dopa therapy inhibited RAS overactivity in young dopamine-depleted rats, but was ineffective in aged rats. The results suggest that dopamine may play an important role in modulating oxidative stress and inflammation in the substantia nigra and striatum via the RAS, which is impaired by aging.
Collapse
|
31
|
Zhang LN, Li JX, Hao L, Sun YJ, Xie YH, Wu SM, Liu L, Chen XL, Gao ZB. Crosstalk between dopamine receptors and the Na⁺/K⁺-ATPase (review). Mol Med Rep 2013; 8:1291-9. [PMID: 24065247 DOI: 10.3892/mmr.2013.1697] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 09/05/2013] [Indexed: 11/06/2022] Open
Abstract
Dopamine (DA) receptors, which belong to the G protein-coupled receptor family, are the target of ~50% of all modern medicinal drugs and constitute a large and diverse class of proteins whose primary function is to transduce extracellular stimuli into intracellular signals. Na+/K+-ATPase (NKA) is ubiquitous and crucial for the maintenance of intracellular ion homeostasis and excitability. Furthermore, it plays a critical role in diverse effects, including clinical cardiotonic and cardioprotective effects, ischemic preconditioning in the brain, natriuresis, lung edema clearance and other processes. NKA regulation is of physiological and pharmacological importance and has species- and tissue-specific variations. The activation of DA receptors regulates NKA expression/activity and trafficking in various tissues and cells, for example in the kidney, lung, intestine, brain, non-pigmented ciliary epithelium and the vascular bed. DA receptor-mediated regulation of NKA mediates a diverse range of cellular responses and includes endocytosis/exocytosis, phosphorylation/dephosphorylation of the α subunit of NKA and multiple signaling pathways, including phosphatidylinositol (PI)-phospholipase C/protein kinase (PK) C, cAMP/PKA, PI3K, adaptor protein 2, tyrosine phosphatase and mitogen-activated protein kinase/extracellular signal-regulated protein kinase. Furthermore, in brain and HEK293T cells, D1 and D2 receptors exist in a complex with NKA. Among D1 and D2 receptors and NKA, regulations are reciprocal, which leads to crosstalk between DA receptors and NKA. In the present study, the current understanding of signaling mechanisms responsible for the crosstalk between DA receptors and NKA, as well as with specific consequent functions, is reviewed.
Collapse
Affiliation(s)
- Li-Nan Zhang
- Department of Pharmacy, College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei 050018, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Renal dopamine receptors, oxidative stress, and hypertension. Int J Mol Sci 2013; 14:17553-72. [PMID: 23985827 PMCID: PMC3794741 DOI: 10.3390/ijms140917553] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/09/2013] [Accepted: 08/12/2013] [Indexed: 12/22/2022] Open
Abstract
Dopamine, which is synthesized in the kidney, independent of renal nerves, plays an important role in the regulation of fluid and electrolyte balance and systemic blood pressure. Lack of any of the five dopamine receptor subtypes (D1R, D2R, D3R, D4R, and D5R) results in hypertension. D1R, D2R, and D5R have been reported to be important in the maintenance of a normal redox balance. In the kidney, the antioxidant effects of these receptors are caused by direct and indirect inhibition of pro-oxidant enzymes, specifically, nicotinamide adenine dinucleotide phosphate, reduced form (NADPH) oxidase, and stimulation of anti-oxidant enzymes, which can also indirectly inhibit NADPH oxidase activity. Thus, stimulation of the D2R increases the expression of endogenous anti-oxidants, such as Parkinson protein 7 (PARK7 or DJ-1), paraoxonase 2 (PON2), and heme oxygenase 2 (HO-2), all of which can inhibit NADPH oxidase activity. The D5R decreases NADPH oxidase activity, via the inhibition of phospholipase D2, and increases the expression of HO-1, another antioxidant. D1R inhibits NADPH oxidase activity via protein kinase A and protein kinase C cross-talk. In this review, we provide an overview of the protective roles of a specific dopamine receptor subtype on renal oxidative stress, the different mechanisms involved in this effect, and the role of oxidative stress and impairment of dopamine receptor function in the hypertension that arises from the genetic ablation of a specific dopamine receptor gene in mice.
Collapse
|
33
|
Dominguez-Meijide A, Villar-Cheda B, Garrido-Gil P, Sierrra-Paredes G, Guerra MJ, Labandeira-Garcia JL. Effect of chronic treatment with angiotensin type 1 receptor antagonists on striatal dopamine levels in normal rats and in a rat model of Parkinson's disease treated with L-DOPA. Neuropharmacology 2013; 76 Pt A:156-68. [PMID: 23973568 DOI: 10.1016/j.neuropharm.2013.07.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/20/2013] [Accepted: 07/17/2013] [Indexed: 01/08/2023]
Abstract
Beneficial effects of angiotensin type-1 receptor (AT1) inhibition have been observed in a number of brain processes mediated by oxidative stress and neuroinflammation, including Parkinson's disease. However, important counterregulatory interactions between dopamine and angiotensin systems have recently been demonstrated in several peripheral tissues, and it is possible that a decrease in dopamine levels due to AT1 inhibition may interfere with neuroprotective strategies. The present experiments involving rats with normal dopaminergic innervation indicate that chronic treatment with the AT1 antagonist candesartan does not significantly affect striatal levels of dopamine, serotonin or metabolites, as does not significantly affect motor behavior, as evaluated by the rotarod test. Interestingly, chronic administration of candesartan to normal rats induced a marked increase in dopamine D1 and a decrease in dopamine D2 receptor expression. In a rat model of Parkinson's disease treated with L-DOPA, no differences in striatal dopamine and serotonin levels were observed between candesartan-treated rats and untreated, which suggests that chronic treatment with candesartan does not significantly affect the process of L-DOPA decarboxylation and dopamine release in Parkinson's disease patients. Candesartan did not induce any differences in the striatal expression of dopamine D1 and D2 and serotonin 5-HT1B receptors in 6ydroxydopamine-lesioned rats treated with L-DOPA. The results suggest that chronic treatment with AT1 antagonists as a neuroprotective strategy does not significantly affect striatal dopamine release or motor behavior. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Antonio Dominguez-Meijide
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Spain
| | | | | | | | | | | |
Collapse
|
34
|
Labandeira-Garcia JL, Rodriguez-Pallares J, Dominguez-Meijide A, Valenzuela R, Villar-Cheda B, Rodríguez-Perez AI. Dopamine-angiotensin interactions in the basal ganglia and their relevance for Parkinson's disease. Mov Disord 2013; 28:1337-42. [PMID: 23925977 DOI: 10.1002/mds.25614] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/29/2013] [Accepted: 06/26/2013] [Indexed: 01/08/2023] Open
Abstract
Renin-angiotensin systems are known to act in many tissues, for example, the blood vessel wall or kidney, where a close interaction between angiotensin and dopamine has been demonstrated. Regulatory interactions between the dopaminergic and renin-angiotensin systems have recently been described in the substantia nigra and striatum. In animal models, dopamine depletion induces compensatory overactivation of the local renin-angiotensin system, which primes microglial responses and neuron vulnerability by activating NADPH-oxidase. Hyperactivation of the local renin-angiotensin system exacerbates the inflammatory microglial response, oxidative stress, and dopaminergic degeneration, all of which are inhibited by angiotensin receptor blockers and inhibitors of angiotensin-converting enzymes. In this review we provide evidence suggesting that the renin-angiotensin system may play an important role in dopamine's mediated neuroinflammation and oxidative stress changes in Parkinson's disease. We suggest that manipulating brain angiotensin may constitute an effective neuroprotective strategy for Parkinson's disease.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW One-third of the world's population has hypertension and it is responsible for almost 50% of deaths from stroke or coronary heart disease. These statistics do not distinguish salt-sensitive from salt-resistant hypertension or include normotensives who are salt-sensitive even though salt sensitivity, independent of blood pressure, is a risk factor for cardiovascular and other diseases, including cancer. This review describes new personalized diagnostic tools for salt sensitivity. RECENT FINDINGS The relationship between salt intake and cardiovascular risk is not linear, but rather fits a J-shaped curve relationship. Thus, a low-salt diet may not be beneficial to everyone and may paradoxically increase blood pressure in some individuals. Current surrogate markers of salt sensitivity are not adequately sensitive or specific. Tests in the urine that could be surrogate markers of salt sensitivity with a quick turn-around time include renal proximal tubule cells, exosomes, and microRNA shed in the urine. SUMMARY Accurate testing of salt sensitivity is not only laborious but also expensive, and with low patient compliance. Patients who have normal blood pressure but are salt-sensitive cannot be diagnosed in an office setting and there are no laboratory tests for salt sensitivity. Urinary surrogate markers for salt sensitivity are being developed.
Collapse
|
36
|
Gildea JJ, Tran HT, Van Sciver RE, Bigler Wang D, Carlson JM, Felder RA. A novel role for c-Myc in G protein-coupled receptor kinase 4 (GRK4) transcriptional regulation in human kidney proximal tubule cells. Hypertension 2013; 61:1021-7. [PMID: 23509080 DOI: 10.1161/hypertensionaha.111.00321] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The G protein-coupled receptor kinase 4 (GRK4) negatively regulates the dopaminergic system by desensitizing the dopamine-1-receptor. The expressional control of GRK4 has not been reported, but here we show that the transcription factor c-Myc binds to the promoter of GRK4 and positively regulates GRK4 protein expression in human renal proximal tubule cells (RPTCs). Addition of phorbol esters to RPTCs not only increased c-Myc binding to the GRK4 promoter but also increased both phospho-c-Myc and GRK4 expression. The phorbol ester-mediated increase in GRK4 expression was completely blocked by the c-Myc inhibitor, 10074-G5, indicating that GRK4 is downstream of phospho-c-Myc. The autocrine production of angiotensin II (Ang II) in RPTCs increased the phosphorylation and activation of c-Myc and subsequently GRK4 expression. 3-Amino-4-thio-butyl sulfonate, an inhibitor of aminopeptidase A, increased RPTC secretion of Ang II. 3-Amino-4-thio-butyl sulfonate or Ang II increased the expression of both phospho-c-Myc and GRK4, which was blocked by 10074-G5. Blockade of the Ang II type 1 receptor with losartan decreased phospho-c-Myc and GRK4 expression. Both inhibition of c-Myc activity and blockade of Ang II type 1 receptor restored the coupling of dopamine-1-receptor to adenylyl cyclase stimulation in uncoupled RPTCs, whereas phorbol esters or Ang II caused the uncoupling of normally coupled RPTCs. We suggest that the Ang II type 1 receptor impairs dopamine-1-receptor function via c-Myc activation of GRK4. This novel pathway may be involved in the increase in blood pressure in hypertension that is mediated by increased activity of the renin-angiotensin system and decreased activity of the renal dopaminergic system.
Collapse
Affiliation(s)
- John J Gildea
- Department of Pathology, University of Virginia Health System, Charlottesville, VA 22908, USA.
| | | | | | | | | | | |
Collapse
|
37
|
A noncanonical postsynaptic transport route for a GPCR belonging to the serotonin receptor family. J Neurosci 2013; 32:17998-8008. [PMID: 23238716 DOI: 10.1523/jneurosci.1804-12.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Postsynaptic receptor trafficking plays an essential role in tuning neurotransmission and signal plasticity and has emerged as a potential therapeutic target in neuropsychiatric disease. Using a novel application of fluorescence recovery after photobleaching in rat hippocampal neurons, we examined transport from the soma to dendrites of seven G-protein-coupled receptors (GPCRs) implicated in mood disorders. Most GPCRs were delivered to dendrites via lateral diffusion, but one GPCR, the serotonin 1B receptor (5-HT(1B)), was delivered to the dendrites in secretory vesicles. Within the dendrites, 5-HT(1B) were stored in a reservoir of accessible vesicles that were recruited to preferential sites in plasma membrane, as observed with superecliptic pHluorin labeling. After membrane recruitment, 5-HT(1B) transport via lateral diffusion and temporal confinement to inhibitory and excitatory synapses was monitored by single particle tracking. These results suggest an alternative mechanism for control of neuronal activity via a GPCR that has been implicated in mood regulation.
Collapse
|
38
|
Gildea JJ, Lahiff DT, Van Sciver RE, Weiss RS, Shah N, McGrath HE, Schoeffel CD, Jose PA, Carey RM, Felder RA. A linear relationship between the ex-vivo sodium mediated expression of two sodium regulatory pathways as a surrogate marker of salt sensitivity of blood pressure in exfoliated human renal proximal tubule cells: the virtual renal biopsy. Clin Chim Acta 2013; 421:236-42. [PMID: 23454474 DOI: 10.1016/j.cca.2013.02.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 01/14/2013] [Accepted: 02/21/2013] [Indexed: 12/30/2022]
Abstract
BACKGROUND Salt sensitivity (SS) of blood pressure (BP) affects 25% of adults, shares comorbidity with hypertension, and has no convenient diagnostic test. We tested the hypothesis that urine-derived exfoliated renal proximal tubule cells (RPTCs) could diagnose the degree of an individual's SS of BP. METHODS Subjects were selected who had their SS of BP determined 5 y prior to this study (salt-sensitive: ≥7 mm Hg increase in mean arterial pressure (MAP) following transition from a random weekly diet of low (10 mmol/day) to high (300 mmol/day) sodium (Na(+)) intake, N=4; inverse salt-sensitive (ISS): ≥7 mm Hg increase in MAP transitioning from a high to low Na(+) diet, N=3, and salt-resistant (SR): <7 mm Hg change in MAP transitioned on either diet, N=5). RPTC responses to 2 independent Na(+) transport pathways were measured. RESULTS There was a negative correlation between the degree of SS and dopamine-1 receptor (D1R) plasma membrane recruitment (y=-0.0107x+0.68 relative fluorescent units (RFU), R(2)=0.88, N=12, P<0.0001) and angiotensin II-stimulated intracellular Ca(++) (y=-0.0016x+0.0336, R(2)=0.7112, P<0.001, N=10) concentration over baseline. CONCLUSIONS Isolating RPTCs from urine provides a personalized cell-based diagnostic test of SS index that offers advantages over a 2-week controlled diet with respect to cost and patient compliance. Furthermore, the linear relationship between the change in MAP and response to 2 Na(+) regulatory pathways suggests that an individual's RPTC response to intracellular Na(+) is personalized and predictive.
Collapse
Affiliation(s)
- John J Gildea
- University of Virginia, Charlottesville, VA 22903, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Azarias G, Kruusmägi M, Connor S, Akkuratov EE, Liu XL, Lyons D, Brismar H, Broberger C, Aperia A. A specific and essential role for Na,K-ATPase α3 in neurons co-expressing α1 and α3. J Biol Chem 2012. [PMID: 23195960 DOI: 10.1074/jbc.m112.425785] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Most neurons co-express two catalytic isoforms of Na,K-ATPase, the ubiquitous α1, and the more selectively expressed α3. Although neurological syndromes are associated with α3 mutations, the specific role of this isoform is not completely understood. Here, we used electrophysiological and Na(+) imaging techniques to study the role of α3 in central nervous system neurons expressing both isoforms. Under basal conditions, selective inhibition of α3 using a low concentration of the cardiac glycoside, ouabain, resulted in a modest increase in intracellular Na(+) concentration ([Na(+)](i)) accompanied by membrane potential depolarization. When neurons were challenged with a large rapid increase in [Na(+)](i), similar to what could be expected following suprathreshold neuronal activity, selective inhibition of α3 almost completely abolished the capacity to restore [Na(+)](i) in soma and dendrite. Recordings of Na,K-ATPase specific current supported the notion that when [Na(+)](i) is elevated in the neuron, α3 is the predominant isoform responsible for rapid extrusion of Na(+). Low concentrations of ouabain were also found to disrupt cortical network oscillations, providing further support for the importance of α3 function in the central nervous system. The α isoforms express a well conserved protein kinase A consensus site, which is structurally associated with an Na(+) binding site. Following activation of protein kinase A, both the α3-dependent current and restoration of dendritic [Na(+)](i) were significantly attenuated, indicating that α3 is a target for phosphorylation and may participate in short term regulation of neuronal function.
Collapse
Affiliation(s)
- Guillaume Azarias
- Department of Women's and Children's Health, Karolinska Institutet, 171 76 Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chugh G, Pokkunuri I, Asghar M. Renal dopamine and angiotensin II receptor signaling in age-related hypertension. Am J Physiol Renal Physiol 2012; 304:F1-7. [PMID: 23097467 DOI: 10.1152/ajprenal.00441.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Kidneys play a vital role in long-term regulation of blood pressure. This is achieved by actions of many renal and nonrenal factors acting on the kidney that help maintain the body's water and electrolyte balance and thus control blood pressure. Several endogenously formed or circulating hormones/peptides, by acting within the kidney, regulate fluid and water homeostasis and blood pressure. Dopamine and angiotensin II are the two key renal factors that, via acting on their receptors and counterregulating each other's function, maintain water and sodium balance. In this review, we provide recent advances in the signaling cascades of these renal receptors, especially at the level of their cross talk, and discuss their roles in blood pressure regulation in the aging process.
Collapse
Affiliation(s)
- Gaurav Chugh
- Heart and Kidney Institute, College of Pharmacy, Univ. of Houston, Houston, TX 77204, USA
| | | | | |
Collapse
|
41
|
Du Z, Wan L, Yan Q, Weinbaum S, Weinstein AM, Wang T. Regulation of glomerulotubular balance: II: impact of angiotensin II on flow-dependent transport. Am J Physiol Renal Physiol 2012; 303:F1507-16. [PMID: 22952281 DOI: 10.1152/ajprenal.00277.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Underlying glomerulotubular balance (GTB) is the impact of axial flow to regulate Na(+) and HCO(3)(-) transport by modulating Na(+)-H(+) exchanger 3 (NHE3) and H-ATPase activity. It is not known whether the cascade of events following a change in flow relies on local angiotensin (ANG II) generation or receptor availability. Mouse tubules were microperfused in vitro at flows of 5 and 20 nl/min, and net fluid (J(v)) and HCO(3)(-) (J(HCO3)) absorption and cell height were measured. Na(+) (J(Na)) and Cl(-) (J(Cl)) absorption and changes in microvillous torque were estimated. Raising flow increased Na(+) and HCO(3)(-) reabsorption but did not change either Cl(-) transport or cell volume. Losartan reduced absolute Na(+) and HCO(3)(-) absorption at both low and high flows but did not affect fractional flow-stimulated transport. Compared with controls, in AT(1a) knockout (KO) mouse tubules, 53% of flow-stimulated Na(+) absorption was abolished, but flow-stimulated HCO(3)(-) absorption was retained at similar levels. The remaining flow-stimulated J(HCO3) was eliminated by the H-ATPase inhibitor bafilomycin. Inhibition of the AT(2) receptor by PD123319 increased both J(Na) and J(HCO3) but did not affect flow-mediated fractional changes. NHE3 expression at the protein level was reduced in AT(1a) KO mice kidneys. We conclude that 1) although the AT(1a) receptor is necessary for flow to impact NHE3, the effect on H(+)-ATPase is independent of AT(1a); 2) the small flow-mediated changes in cell volume suggest a coordinate flow effect on both luminal and basolateral transporters; and 3) there is no evidence of flow-dependent Cl(-) transport, and thus no evidence for convective paracellular Cl(-) transport in mouse tubules.
Collapse
Affiliation(s)
- Zhaopeng Du
- Dept. of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06520-8026, USA
| | | | | | | | | | | |
Collapse
|
42
|
Rodriguez-Pallares J, Parga JA, Joglar B, Guerra MJ, Labandeira-Garcia JL. Mitochondrial ATP-sensitive potassium channels enhance angiotensin-induced oxidative damage and dopaminergic neuron degeneration. Relevance for aging-associated susceptibility to Parkinson's disease. AGE (DORDRECHT, NETHERLANDS) 2012; 34:863-880. [PMID: 21713375 PMCID: PMC3682060 DOI: 10.1007/s11357-011-9284-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 06/15/2011] [Indexed: 05/31/2023]
Abstract
Recent studies have shown that renin-angiotensin system overactivation is involved in the aging process in several tissues as well as in longevity and aging-related degenerative diseases by increasing oxidative damage and inflammation. We have recently shown that angiotensin II enhances dopaminergic degeneration by increasing levels of reactive oxygen species and neuroinflammation, and that there is an aging-related increase in angiotensin II activity in the substantia nigra in rats, which may constitute a major factor in the increased risk of Parkinson's disease with aging. The mechanisms involved in the above mentioned effects and particularly a potential angiotensin-mitochondria interaction have not been clarified. The present study revealed that activation of mitochondrial ATP-sensitive potassium channels [mitoK(ATP)] may play a major role in the angiotensin II-induced effects on aging and neurodegeneration. Inhibition of mitoK(ATP) channels with 5-hydroxydecanoic acid inhibited the increase in dopaminergic cell death induced by angiotensin II, as well as the increase in superoxide/superoxide-derived reactive oxygen species levels and the angiotensin II-induced decrease in the mitochondrial inner membrane potential in cultured dopaminergic neurons. The present study provides data for considering brain renin-angiotensin system and mitoK(ATP) channels as potential targets for protective therapy in aging-associated diseases such as Parkinson's disease.
Collapse
Affiliation(s)
- Jannette Rodriguez-Pallares
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Center on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Juan Andres Parga
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Center on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Belen Joglar
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Center on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Maria Jose Guerra
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Center on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| | - Jose Luis Labandeira-Garcia
- />Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- />Networking Research Center on Neurodegenerative Diseases (CIBERNED), Santiago de Compostela, Spain
| |
Collapse
|
43
|
Aperia A. 2011 Homer Smith Award: To serve and protect: classic and novel roles for Na+, K+ -adenosine triphosphatase. J Am Soc Nephrol 2012; 23:1283-90. [PMID: 22745476 DOI: 10.1681/asn.2012010102] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The ability of cells to maintain sharp ion gradients across their membranes is the foundation for the molecular transport and electrical excitability. Across animal species and cell types, Na(+),K(+)-adenosine triphosphatase (ATPase) is arguably the most powerful contributor to this phenomenon. By producing a steep concentration difference of sodium and potassium between the intracellular and extracellular milieu, Na(+),K(+)-ATPase in the tubules provides the driving force for renal sodium reabsorption. Pump activity is downregulated by natriuretic hormones, such as dopamine, and is upregulated by antinatriuretic hormones, such as angiotensin. In the past decade, studies have revealed a novel and surprising role: that Na(+),K(+)-ATPase is a transducer of signals from extracellular to intracellular compartments. The signaling function of Na(+),K(+)-ATPase is activated by ouabain, a mammalian steroid hormone, at far lower concentrations than those that inhibit pump activity. By promoting growth and inhibiting apoptosis, activation of Na(+),K(+)-ATPase exerts tissue-protective effects. Ouabain-stimulated Na(+),K(+)-ATPase signaling has recently shown clinical promise by protecting the malnourished embryonic kidney from adverse developmental programming. A deeper understanding of the tissue-protective role of Na(+),K(+)-ATPase signaling and the regulation of Na(+),K(+)-ATPase pumping activity is of fundamental importance for the understanding and treatment of kidney diseases and kidney-related hypertension.
Collapse
Affiliation(s)
- Anita Aperia
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Q2-09 SE-171 76 Stockholm, Sweden.
| |
Collapse
|
44
|
Du Z, Yan Q, Wan L, Weinbaum S, Weinstein AM, Wang T. Regulation of glomerulotubular balance. I. Impact of dopamine on flow-dependent transport. Am J Physiol Renal Physiol 2012; 303:F386-95. [PMID: 22552936 DOI: 10.1152/ajprenal.00531.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
In response to volume expansion, locally generated dopamine decreases proximal tubule reabsorption by reducing both Na/H-exchanger 3 (NHE3) and Na-K-ATPase activity. We have previously demonstrated that mouse proximal tubules in vitro respond to changes in luminal flow with proportional changes in Na(+) and HCO(3)(-) reabsorption and have suggested that this observation underlies glomerulotubular balance. In the present work, we investigate the impact of dopamine on the sensitivity of reabsorptive fluxes to changes in luminal flow. Mouse proximal tubules were microperfused in vitro at low and high flow rates, and volume and HCO(3)(-) reabsorption (J(v) and J(HCO3)) were measured, while Na(+) and Cl(-) reabsorption (J(Na) and J(Cl)) were estimated. Raising luminal flow increased J(v), J(Na), and J(HCO3) but did not change J(Cl). Luminal dopamine did not change J(v), J(Na), and J(HCO3) at low flow rates but completely abolished the increments of Na(+) absorption by flow and partially inhibited the flow-stimulated HCO(3)(-) absorption. The remaining flow-stimulated HCO(3)(-) absorption was completely abolished by bafilomycin. The DA1 receptor blocker SCH23390 and the PKA inhibitor H89 blocked the effect of exogenous dopamine and produced a two to threefold increase in the sensitivity of proximal Na(+) reabsorption to luminal flow rate. Under the variety of perfusion conditions, changes in cell volume were small and did not always parallel changes in Na(+) transport. We conclude that 1) dopamine inhibits flow-stimulated NHE3 activity by activation of the DA1 receptor via a PKA-mediated mechanism; 2) dopamine has no effect on flow-stimulated H-ATPase activity; 3) there is no evidence of flow stimulation of Cl(-) reabsorption; and 4) the impact of dopamine is a coordinated modulation of both luminal and peritubular Na(+) transporters.
Collapse
Affiliation(s)
- Zhaopeng Du
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06520-8026, USA
| | | | | | | | | | | |
Collapse
|
45
|
Garrido-Gil P, Valenzuela R, Villar-Cheda B, Lanciego JL, Labandeira-Garcia JL. Expression of angiotensinogen and receptors for angiotensin and prorenin in the monkey and human substantia nigra: an intracellular renin-angiotensin system in the nigra. Brain Struct Funct 2012; 218:373-88. [PMID: 22407459 PMCID: PMC3580133 DOI: 10.1007/s00429-012-0402-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 02/22/2012] [Indexed: 02/07/2023]
Abstract
We have previously obtained in rodents a considerable amount of data suggesting a major role for the brain renin–angiotensin system (RAS) in dopaminergic neuron degeneration and potentially in Parkinson’s disease. However, the presence of a local RAS has not been demonstrated in the monkey or the human substantia nigra compacta (SNc). The present study demonstrates the presence of major RAS components in dopaminergic neurons, astrocytes and microglia in both the monkey and the human SNc. Angiotensin type 1 and 2 and renin–prorenin receptors were located at the surface of dopaminergic neurons and glial cells, as expected for a tissular RAS. However, angiotensinogen and receptors for angiotensin and renin–prorenin were also observed at the cytoplasm and nuclear level, which suggests the presence of an intracrine or intracellular RAS in monkey and human SNc. Although astrocytes and microglia were labeled for angiotensin and prorenin receptors in the normal SNc, most glial cells appeared less immunoreactive than the dopaminergic neurons. However, our previous studies in rodent models of PD and studies in other animal models of brain diseases suggest that the RAS activity is significantly upregulated in glial cells in pathological conditions. The present results together with our previous findings in rodents suggest a major role for the nigral RAS in the normal functioning of the dopaminergic neurons, and in the progression of the dopaminergic degeneration.
Collapse
Affiliation(s)
- Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Faculty of Medicine, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | | | | | | | | |
Collapse
|
46
|
Li D, Scott L, Crambert S, Zelenin S, Eklöf AC, Di Ciano L, Ibarra F, Aperia A. Binding of losartan to angiotensin AT1 receptors increases dopamine D1 receptor activation. J Am Soc Nephrol 2011; 23:421-8. [PMID: 22193384 DOI: 10.1681/asn.2011040344] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Signaling through both angiotensin AT1 receptors (AT1R) and dopamine D1 receptors (D1R) modulates renal sodium excretion and arterial BP. AT1R and D1R form heterodimers, but whether treatment with AT1R antagonists functionally modifies D1R via allosterism is unknown. In this study, the AT1R antagonist losartan strengthened the interaction between AT1R and D1R and increased expression of D1R on the plasma membrane in vitro. In rat proximal tubule cells that express endogenous AT1R and D1R, losartan increased cAMP generation. Losartan increased cAMP in HEK 293a cells transfected with both AT1R and D1R, but it did not increase cAMP in cells transfected with either receptor alone, suggesting that losartan induces D1R activation. Furthermore, losartan did not increase cAMP in HEK 293a cells expressing AT1R and mutant S397/S398A D1R, which disrupts the physical interaction between AT1R and D1R. In vivo, administration of a D1R antagonist significantly attenuated the antihypertensive effect of losartan in rats with renal hypertension. Taken together, these data imply that losartan might exert its antihypertensive effect both by inhibiting AT1R signaling and by enhancing D1R signaling.
Collapse
Affiliation(s)
- Dong Li
- Department of Women's and Children's Health, Karolinska University Hospital, Q2:09, S-171 76 Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Asghar M, Tayebati SK, Lokhandwala MF, Hussain T. Potential dopamine-1 receptor stimulation in hypertension management. Curr Hypertens Rep 2011; 13:294-302. [PMID: 21633929 DOI: 10.1007/s11906-011-0211-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The role of dopamine receptors in blood pressure regulation is well established. Genetic ablation of both dopamine D1-like receptor subtypes (D1, D5) and D2-like receptor subtypes (D2, D3, D4) results in a hypertensive phenotype in mice. This review focuses on the dopamine D1-like receptor subtypes D1 and D5 (especially D1 receptors), as they play a major role in regulating sodium homeostasis and blood pressure. Studies mostly describing the role of renal dopamine D1-like receptors are included, as the kidneys play a pivotal role in the maintenance of sodium homeostasis and the long-term regulation of blood pressure. We also attempt to describe the interaction between D1-like receptors and other proteins, especially angiotensin II type 1 and type 2 receptors, which are involved in the maintenance of sodium homeostasis and blood pressure. Finally, we discuss a new concept of renal D1 receptor regulation in hypertension that involves oxidative stress mechanisms.
Collapse
Affiliation(s)
- Mohammad Asghar
- Heart and Kidney Institute, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| | | | | | | |
Collapse
|
48
|
Rodriguez-Perez AI, Valenzuela R, Joglar B, Garrido-Gil P, Guerra MJ, Labandeira-Garcia JL. Renin angiotensin system and gender differences in dopaminergic degeneration. Mol Neurodegener 2011; 6:58. [PMID: 21846363 PMCID: PMC3169490 DOI: 10.1186/1750-1326-6-58] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 08/16/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are sex differences in dopaminergic degeneration. Men are approximately two times as likely as premenopausal women of the same age to develop Parkinson's disease (PD). It has been shown that the local renin angiotensin system (RAS) plays a prominent role in sex differences in the development of chronic renal and cardiovascular diseases, and there is a local RAS in the substantia nigra and dopaminergic cell loss is enhanced by angiotensin via type 1 (AT1) receptors. RESULTS In the present study, we observed that intrastriatal injection of 6-hydroxydopamine induced a marked loss of dopaminergic neurons in the substantia nigra of male rats, which was significantly higher than the loss induced in ovariectomized female rats given estrogen implants (i.e. rats with estrogen). However, the loss of dopaminergic neurons was significantly lower in male rats treated with the AT1 antagonist candesartan, and similar to that observed in female rats with estrogen. The involvement of the RAS in gender differences in dopaminergic degeneration was confirmed with AT1a-null mice lesioned with the dopaminergic neurotoxin MPTP. Significantly higher expression of AT1 receptors, angiotensin converting enzyme activity, and NADPH-oxidase complex activity, and much lower levels of AT2 receptors were observed in male rats than in female rats with estrogen. CONCLUSIONS The results suggest that brain RAS plays a major role in the increased risk of developing PD in men, and that manipulation of brain RAS may be an efficient approach for neuroprotective treatment of PD in men, without the feminizing effects of estrogen.
Collapse
Affiliation(s)
- Ana I Rodriguez-Perez
- Department of Morphological Sciences, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Santiago de Compostela, Santiago de Compostela, E-15782 Spain
| | - Rita Valenzuela
- Department of Morphological Sciences, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Santiago de Compostela, Santiago de Compostela, E-15782 Spain
| | - Belen Joglar
- Department of Morphological Sciences, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Santiago de Compostela, Santiago de Compostela, E-15782 Spain
| | - Pablo Garrido-Gil
- Department of Morphological Sciences, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Santiago de Compostela, Santiago de Compostela, E-15782 Spain
| | - Maria J Guerra
- Department of Morphological Sciences, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Santiago de Compostela, Santiago de Compostela, E-15782 Spain
| | - Jose L Labandeira-Garcia
- Department of Morphological Sciences, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Santiago de Compostela, Santiago de Compostela, E-15782 Spain
| |
Collapse
|
49
|
Abstract
Dopamine is an important regulator of systemic blood pressure via multiple mechanisms. It affects fluid and electrolyte balance by its actions on renal hemodynamics and epithelial ion and water transport and by regulation of hormones and humoral agents. The kidney synthesizes dopamine from circulating or filtered L-DOPA independently from innervation. The major determinants of the renal tubular synthesis/release of dopamine are probably sodium intake and intracellular sodium. Dopamine exerts its actions via two families of cell surface receptors, D1-like receptors comprising D1R and D5R, and D2-like receptors comprising D2R, D3R, and D4R, and by interactions with other G protein-coupled receptors. D1-like receptors are linked to vasodilation, while the effect of D2-like receptors on the vasculature is variable and probably dependent upon the state of nerve activity. Dopamine secreted into the tubular lumen acts mainly via D1-like receptors in an autocrine/paracrine manner to regulate ion transport in the proximal and distal nephron. These effects are mediated mainly by tubular mechanisms and augmented by hemodynamic mechanisms. The natriuretic effect of D1-like receptors is caused by inhibition of ion transport in the apical and basolateral membranes. D2-like receptors participate in the inhibition of ion transport during conditions of euvolemia and moderate volume expansion. Dopamine also controls ion transport and blood pressure by regulating the production of reactive oxygen species and the inflammatory response. Essential hypertension is associated with abnormalities in dopamine production, receptor number, and/or posttranslational modification.
Collapse
Affiliation(s)
- Ines Armando
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| | - Van Anthony M. Villar
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| | - Pedro A. Jose
- Children’s National Medical Center—Center for Molecular Physiology Research, Washington, District of Columbia
| |
Collapse
|
50
|
Abstract
The assessment of salt sensitivity of blood pressure is difficult because of the lack of universal consensus on definition. Regardless of the variability in the definition of salt sensitivity, increased salt intake, independent of the actual level of blood pressure, is also a risk factor for cardiovascular morbidity and mortality and kidney disease. A modest reduction in salt intake results in an immediate decrease in blood pressure, with long-term beneficial consequences. However, some have suggested that dietary sodium restriction may not be beneficial to everyone. Thus, there is a need to distinguish salt-sensitive from salt-resistant individuals, but it has been difficult to do so with phenotypic studies. Therefore, there is a need to determine the genes that are involved in salt sensitivity. This review focuses on genes associated with salt sensitivity, with emphasis on the variants associated with salt sensitivity in humans that are not due to monogenic causes. Special emphasis is given to gene variants associated with salt sensitivity whose protein products interfere with cell function and increase blood pressure in transgenic mice.
Collapse
Affiliation(s)
- Hironobu Sanada
- Division of Health Science Research, Fukushima Welfare Federation of Agricultural Cooperatives, Fukushima, Japan.
| | | | | |
Collapse
|