1
|
Crispo JAG, Farhat N, Fortin Y, Perez-Lloret S, Sikora L, Morgan RL, Habash M, Gogna P, Kelly SE, Elliott J, Kohen DE, Bjerre LM, Mattison DR, Hessian RC, Willis AW, Krewski D. Non-Ergot Dopamine Agonists and the Risk of Heart Failure and Other Adverse Cardiovascular Reactions in Parkinson's Disease. Brain Sci 2024; 14:776. [PMID: 39199470 PMCID: PMC11352331 DOI: 10.3390/brainsci14080776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Reports suggest possible risks of adverse cardiovascular reactions, including heart failure, associated with non-ergot dopamine agonist (DA) use in Parkinson's disease (PD). The objectives of our review were to evaluate the risk of heart failure and other adverse cardiovascular reactions in PD patients who received a non-ergot DA compared with other anti-PD pharmacological interventions, placebo, or no intervention. Studies were identified via searches of six bibliographic databases. Randomized controlled trials (RCTs) and non-randomized studies (NRS) were eligible for study inclusion. Random-effect meta-analyses were performed to estimate adverse cardiovascular reaction risks. Quality of evidence was assessed using GRADE. In total, forty-four studies (thirty-six RCTs and eight NRS) satisfied our inclusion criteria. A single RCT found no significant difference in the risk of heart failure with ropinirole compared with bromocriptine (odds ratio (OR) 0.39, 95% confidence interval (CI) 0.07 to 2.04; low certainty). Conversely, three case-control studies reported a risk of heart failure with non-ergot DA treatment. The quality of evidence for the risk of heart failure was judged as low or very low. Findings suggest that non-ergot DA use may be associated with adverse cardiovascular outcomes, including heart failure. Studies are needed to better understand cardiovascular risks associated with PD treatment.
Collapse
Affiliation(s)
- James A. G. Crispo
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Human Sciences Division, NOSM University, Sudbury, ON P3E 2C6, Canada
| | - Nawal Farhat
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| | - Yannick Fortin
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| | - Santiago Perez-Lloret
- Observatorio de Salud, Pontificia Universidad Católica Argentina, Consejo de Investigaciones Científicas y Técnicas (UCA-CONICET), Buenos Aires C1107AAZ, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires C1121ABG, Argentina
| | - Lindsey Sikora
- Health Sciences Library, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Rebecca L. Morgan
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Mara Habash
- Aboriginal Cancer Control Unit, Cancer Care Ontario, Toronto, ON M5G 2L7, Canada
| | - Priyanka Gogna
- Department of Public Health Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Shannon E. Kelly
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- Cardiovascular Research Methods Centre, University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Jesse Elliott
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| | - Dafna E. Kohen
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
| | - Lise M. Bjerre
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- Department of Family Medicine, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- Institut du Savoir Montfort, Ottawa, ON K1K 0T2, Canada
| | - Donald R. Mattison
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
| | - Renée C. Hessian
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, ON K1Y 4W7, Canada
| | - Allison W. Willis
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, Translational Center of Excellence for Neuroepidemiology and Neurological Outcomes Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Krewski
- McLaughlin Centre for Population Health Risk Assessment, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON K1G 5Z3, Canada
- Risk Sciences International, Ottawa, ON K1P 5J6, Canada
| |
Collapse
|
2
|
Labandeira-Garcia JL, Labandeira CM, Guerra MJ, Rodriguez-Perez AI. The role of the brain renin-angiotensin system in Parkinson´s disease. Transl Neurodegener 2024; 13:22. [PMID: 38622720 PMCID: PMC11017622 DOI: 10.1186/s40035-024-00410-3] [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: 12/13/2023] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
The renin-angiotensin system (RAS) was classically considered a circulating hormonal system that regulates blood pressure. However, different tissues and organs, including the brain, have a local paracrine RAS. Mutual regulation between the dopaminergic system and RAS has been observed in several tissues. Dysregulation of these interactions leads to renal and cardiovascular diseases, as well as progression of dopaminergic neuron degeneration in a major brain center of dopamine/angiotensin interaction such as the nigrostriatal system. A decrease in the dopaminergic function induces upregulation of the angiotensin type-1 (AT1) receptor activity, leading to recovery of dopamine levels. However, AT1 receptor overactivity in dopaminergic neurons and microglial cells upregulates the cellular NADPH-oxidase-superoxide axis and Ca2+ release, which mediate several key events in oxidative stress, neuroinflammation, and α-synuclein aggregation, involved in Parkinson's disease (PD) pathogenesis. An intraneuronal antioxidative/anti-inflammatory RAS counteracts the effects of the pro-oxidative AT1 receptor overactivity. Consistent with this, an imbalance in RAS activity towards the pro-oxidative/pro-inflammatory AT1 receptor axis has been observed in the substantia nigra and striatum of several animal models of high vulnerability to dopaminergic degeneration. Interestingly, autoantibodies against angiotensin-converting enzyme 2 and AT1 receptors are increased in PD models and PD patients and contribute to blood-brain barrier (BBB) dysregulation and nigrostriatal pro-inflammatory RAS upregulation. Therapeutic strategies addressed to the modulation of brain RAS, by AT1 receptor blockers (ARBs) and/or activation of the antioxidative axis (AT2, Mas receptors), may be neuroprotective for individuals with a high risk of developing PD or in prodromal stages of PD to reduce progression of the disease.
Collapse
Affiliation(s)
- Jose Luis Labandeira-Garcia
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | | | - Maria J Guerra
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Cellular and Molecular Neurobiology of Parkinson´S Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain.
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
3
|
Moore SC, Vaz de Castro PAS, Yaqub D, Jose PA, Armando I. Anti-Inflammatory Effects of Peripheral Dopamine. Int J Mol Sci 2023; 24:13816. [PMID: 37762126 PMCID: PMC10530375 DOI: 10.3390/ijms241813816] [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: 05/04/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Dopamine is synthesized in the nervous system where it acts as a neurotransmitter. Dopamine is also synthesized in a number of peripheral organs as well as in several types of cells and has organ-specific functions and, as demonstrated more recently, is involved in the regulation of the immune response and inflammatory reaction. In particular, the renal dopaminergic system is very important in the regulation of sodium transport and blood pressure and is particularly sensitive to stimuli that cause oxidative stress and inflammation. This review is focused on how dopamine is synthesized in organs and tissues and the mechanisms by which dopamine and its receptors exert their effects on the inflammatory response.
Collapse
Affiliation(s)
| | | | | | | | - Ines Armando
- Division of Kidney Diseases and Hypertension, Department of Medicine, The George Washington School of Medicine and Health Sciences, Washington, DC 20037, USA; (S.C.M.); (P.A.S.V.d.C.); (D.Y.); (P.A.J.)
| |
Collapse
|
4
|
Saccharomyces boulardii exerts renoprotection by modulating oxidative stress, renin angiotensin system and uropathogenic microbiota in a murine model of diabetes. Life Sci 2022; 301:120616. [PMID: 35533758 DOI: 10.1016/j.lfs.2022.120616] [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: 12/15/2021] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 11/22/2022]
Abstract
AIMS We aimed to investigate whether Saccharomyces boulardii strain might exert renoprotective effects by modulating renal renin angiotensin system, oxidative stress and intestinal microbiota in streptozotocin-diabetic mice. MAIN METHODS Thirty-six C57BL/6 male mice were divided into four groups: control (C), control + probiotic (CP), diabetes (D), diabetes + probiotic (DP). Diabetes was induced by one intraperitoneal injection of streptozotocin and Saccharomyces boulardii was administered by oral gavage for 8 weeks. Blood glucose, albuminuria and urinary volume were measured. Renal levels of angiotensin peptides (angiotensin I, II and 1-7) and the activities of angiotensin-converting enzyme (ACE) and ACE2 were determined, besides that, renal morphology, serotonin and dopamine levels and also microbiota composition were analyzed. KEY FINDINGS Probiotics significantly increased C-peptide secretion and reduced blood glucose of diabetic animals. Saccharomyces boulardii also improved renal antioxidant defense, restored serotonin and dopamine concentration, and activated the renin-angiotensin system (RAS) vasodilator and antifibrotic axis. The modulation of these markers was associated with a beneficial impact on glomerular structure and renal function of diabetic treated animals. The phenotypic changes induced by Saccharomyces boulardii were also related to modulation of intestinal microbiota, evidenced by the decreased abundance of Proteus and Escherichia-Shigella, considered diabetic nephropathy biomarkers. SIGNIFICANCE Therefore, probiotic administration to streptozotocin-induced diabetic mice improves kidney structure and function in a murine model and might represent a reasonable strategy to counteract nephropathy-associated maladaptive responses in diabetes.
Collapse
|
5
|
Quijano A, Diaz-Ruiz C, Lopez-Lopez A, Villar-Cheda B, Muñoz A, Rodriguez-Perez AI, Labandeira-Garcia JL. Angiotensin Type-1 Receptor Inhibition Reduces NLRP3 Inflammasome Upregulation Induced by Aging and Neurodegeneration in the Substantia Nigra of Male Rodents and Primary Mesencephalic Cultures. Antioxidants (Basel) 2022; 11:antiox11020329. [PMID: 35204211 PMCID: PMC8868290 DOI: 10.3390/antiox11020329] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 12/17/2022] Open
Abstract
The tissue renin–angiotensin system (RAS) has been shown to be involved in prooxidative and proinflammatory changes observed in aging and aging-related diseases such as dopaminergic degeneration in Parkinson’s disease (PD). We studied the activation of the NLRP3 inflammasome in the substantia nigra with aging and early stages of dopaminergic degeneration in PD models and, particularly, if the brain RAS, via its prooxidative proinflammatory angiotensin II (AngII) type 1 (AT1) receptors, mediates the inflammasome activation. Nigras from aged rats and mice and 6-hydroxydopamine PD models showed upregulation in transcription of inflammasome-related components (NLRP3, pro-IL1β and pro-IL18) and IL1β and IL18 protein levels, which was inhibited by the AT1 receptor antagonist candesartan. The role of the AngII/AT1 axis in inflammasome activation was further confirmed in rats intraventricularly injected with AngII, and in primary mesencephalic cultures treated with 6-hydroxydopamine, which showed inflammasome activation that was blocked by candesartan. Observations in the nigra of young and aged AT1 and AT2 knockout mice confirmed the major role of AT1 receptors in nigral inflammasome activation. In conclusion, the inflammasome is upregulated by aging and dopaminergic degeneration in the substantia nigra, possibly related with a decrease in dopamine levels, and it is mediated by the AngII/AT1 axis.
Collapse
Affiliation(s)
- Aloia Quijano
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
| | - Carmen Diaz-Ruiz
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Andrea Lopez-Lopez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Begoña Villar-Cheda
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Ana Muñoz
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Ana I. Rodriguez-Perez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
| | - Jose L. Labandeira-Garcia
- Laboratory of Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Department of Morphological Sciences, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; (A.Q.); (C.D.-R.); (A.L.-L.); (B.V.-C.); (A.M.); (A.I.R.-P.)
- Networking Research Center on Neurodegenerative Diseases (CiberNed), 28031 Madrid, Spain
- Correspondence: ; Tel.: +34-881-812223
| |
Collapse
|
6
|
Shao L, Ma Y, Fang Q, Huang Z, Wan S, Wang J, Yang L. Role of protein phosphatase 2A in kidney disease (Review). Exp Ther Med 2021; 22:1236. [PMID: 34539832 PMCID: PMC8438693 DOI: 10.3892/etm.2021.10671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Kidney disease affects millions of people worldwide and is a financial burden on the healthcare system. Protein phosphatase 2A (PP2A), which is involved in renal development and the function of ion-transport proteins, aquaporin-2 and podocytes, is likely to serve an important role in renal processes. PP2A is associated with the pathogenesis of a variety of different kidney diseases including podocyte injury, inflammation, tumors and chronic kidney disease. The current review aimed to discuss the structure and function of PP2A subunits in the context of kidney diseases. How dysregulation of PP2A in the kidneys causes podocyte death and the inactivation of PP2A in renal carcinoma tissues is discussed. Inhibition of PP2A activity prevents epithelial-mesenchymal transition and attenuates renal fibrosis, creating a favorable inflammatory microenvironment and promoting the initiation and progression of tumor pathogenesis. The current review also indicates that PP2A serves an important role in protection against renal inflammation. Understanding the detailed mechanisms of PP2A provides information that can be utilized in the design and application of novel therapeutics for the treatment and prevention of renal diseases.
Collapse
Affiliation(s)
- Lishi Shao
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Yiqun Ma
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Qixiang Fang
- Department of Urology, The First Affiliated Hospital of the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, P.R. China
| | - Ziye Huang
- Department of Urology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Shanshan Wan
- Department of Radiology, Yunnan Kun-Gang Hospital, Anning, Yunnan 650300, P.R. China
| | - Jiaping Wang
- Department of Radiology, Kunming Medical University and The Second Affiliated Hospital, Kunming, Yunnan 650500, P.R. China
| | - Li Yang
- Department of Anatomy, Histology and Embryology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
7
|
Garrido-Gil P, Rodriguez-Perez AI, Lage L, Labandeira-Garcia JL. Estrogen Deficiency and Colonic Function: Surgical Menopause and Sex Differences in Angiotensin and Dopamine Receptor Interaction. J Gerontol A Biol Sci Med Sci 2021; 76:1533-1541. [PMID: 32991714 DOI: 10.1093/gerona/glaa244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Indexed: 01/02/2025] Open
Abstract
The physiopathological mechanisms that regulate menopausal and sex differences in colonic transit, inflammatory processes, and efficacy of treatments have not been clarified. The dopaminergic system and renin-angiotensin system coexist in the gut and regulate different processes such as motility, absorption/secretion, and inflammation. We investigated the changes in expression of major angiotensin and dopamine receptors in the colon of male, female, and ovariectomized female mice. Possible interaction between both systems was investigated using male and female mice deficient (ko) for major angiotensin and dopamine receptors. In wild-type mice, colonic tissue from females showed lower angiotensin type 1/angiotensin type 2 ratio (an index of pro-inflammatory/anti-inflammatory renin-angiotensin system balance), lower dopamine D1 and D2 receptor expression, and lower levels of pro-inflammatory and pro-oxidative markers relative to males. Interestingly, ovariectomy increased the expression of pro-inflammatory angiotensin type 1 receptor expression and decreased anti-inflammatory angiotensin type 2 receptor expression, increased D1 and D2 receptor expression, and increased the levels of pro-inflammatory and pro-oxidative markers. Ovariectomy-induced changes were blocked by estrogen replacement. The present results suggest a mutual regulation between colonic angiotensin and dopamine receptors and sex differences in this mutual regulation. Estrogen regulates changes in both angiotensin and dopamine receptor expression, which may be involved in sex- and surgical menopause-related effects on gut motility, permeability, and vulnerability to inflammatory processes.
Collapse
Affiliation(s)
- Pablo Garrido-Gil
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Lucia Lage
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Spain
| | - Jose L Labandeira-Garcia
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), IDIS, University of Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
8
|
Angiotensin II induces cognitive decline and anxiety-like behavior via disturbing pattern of theta-gamma oscillations. Brain Res Bull 2021; 174:84-91. [PMID: 34090935 DOI: 10.1016/j.brainresbull.2021.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/13/2021] [Accepted: 06/01/2021] [Indexed: 01/11/2023]
Abstract
Hypertension is the most common chronic disease accompanied by cognitive decline and anxiety-like behavior. Angiotensin II (Ang II) induces hypertension by activating angiotensin II receptor subtype 1 (AT1R). The purpose of the study was to examine the potential underlying mechanism of alterations in cognition and anxiety-like behavior induced by Ang II. Adult C57 mice were intraperitoneal injected with either 1 mg/kg/d Ang II or saline individually for 14 consecutive days. Ang II resulted in cognitive decline and anxious like behavior in C57 mice. Moreover, Ang II disturbed bidirectional synaptic plasticity and neural oscillation coupling between high theta and gamma on PP (perforant pathway)-DG (dentate gyrus) pathway. In addition, Ang II decreased the expression of N-methyl-d-aspartate receptor (NR) 2A and NR 2B and increased the expression of GABAAR α1. The data suggest that Ang II disturb neural oscillations via altering excitatory and inhibitory (E/I) balance and eventually damage cognition and anxiety-like behavior in mice.
Collapse
|
9
|
Labandeira-Garcia JL, Valenzuela R, Costa-Besada MA, Villar-Cheda B, Rodriguez-Perez AI. The intracellular renin-angiotensin system: Friend or foe. Some light from the dopaminergic neurons. Prog Neurobiol 2020; 199:101919. [PMID: 33039415 PMCID: PMC7543790 DOI: 10.1016/j.pneurobio.2020.101919] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/20/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) is one of the oldest hormone systems in vertebrate phylogeny. RAS was initially related to regulation of blood pressure and sodium and water homeostasis. However, local or paracrine RAS were later identified in many tissues, including brain, and play a major role in their physiology and pathophysiology. In addition, a major component, ACE2, is the entry receptor for SARS-CoV-2. Overactivation of tissue RAS leads several oxidative stress and inflammatory processes involved in aging-related degenerative changes. In addition, a third level of RAS, the intracellular or intracrine RAS (iRAS), with still unclear functions, has been observed. The possible interaction between the intracellular and extracellular RAS, and particularly the possible deleterious or beneficial effects of the iRAS activation are controversial. The dopaminergic system is particularly interesting to investigate the RAS as important functional interactions between dopamine and RAS have been observed in the brain and several peripheral tissues. Our recent observations in mitochondria and nucleus of dopaminergic neurons may clarify the role of the iRAS. This may be important for the developing of new therapeutic strategies, since the effects on both extracellular and intracellular RAS must be taken into account, and perhaps better understanding of COVID-19 cell mechanisms.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain.
| | - Rita Valenzuela
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Maria A Costa-Besada
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Begoña Villar-Cheda
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| | - Ana I Rodriguez-Perez
- Laboratory of Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain; Networking Research Center on Neurodegenerative Diseases (CiberNed), Madrid, Spain
| |
Collapse
|
10
|
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: 0.4] [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
|
11
|
Matt SM, Gaskill PJ. Where Is Dopamine and how do Immune Cells See it?: Dopamine-Mediated Immune Cell Function in Health and Disease. J Neuroimmune Pharmacol 2020; 15:114-164. [PMID: 31077015 PMCID: PMC6842680 DOI: 10.1007/s11481-019-09851-4] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/07/2019] [Indexed: 02/07/2023]
Abstract
Dopamine is well recognized as a neurotransmitter in the brain, and regulates critical functions in a variety of peripheral systems. Growing research has also shown that dopamine acts as an important regulator of immune function. Many immune cells express dopamine receptors and other dopamine related proteins, enabling them to actively respond to dopamine and suggesting that dopaminergic immunoregulation is an important part of proper immune function. A detailed understanding of the physiological concentrations of dopamine in specific regions of the human body, particularly in peripheral systems, is critical to the development of hypotheses and experiments examining the effects of physiologically relevant dopamine concentrations on immune cells. Unfortunately, the dopamine concentrations to which these immune cells would be exposed in different anatomical regions are not clear. To address this issue, this comprehensive review details the current information regarding concentrations of dopamine found in both the central nervous system and in many regions of the periphery. In addition, we discuss the immune cells present in each region, and how these could interact with dopamine in each compartment described. Finally, the review briefly addresses how changes in these dopamine concentrations could influence immune cell dysfunction in several disease states including Parkinson's disease, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, as well as the collection of pathologies, cognitive and motor symptoms associated with HIV infection in the central nervous system, known as NeuroHIV. These data will improve our understanding of the interactions between the dopaminergic and immune systems during both homeostatic function and in disease, clarify the effects of existing dopaminergic drugs and promote the creation of new therapeutic strategies based on manipulating immune function through dopaminergic signaling. Graphical Abstract.
Collapse
Affiliation(s)
- S M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
12
|
Leipziger J, Praetorius H. Renal Autocrine and Paracrine Signaling: A Story of Self-protection. Physiol Rev 2020; 100:1229-1289. [PMID: 31999508 DOI: 10.1152/physrev.00014.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autocrine and paracrine signaling in the kidney adds an extra level of diversity and complexity to renal physiology. The extensive scientific production on the topic precludes easy understanding of the fundamental purpose of the vast number of molecules and systems that influence the renal function. This systematic review provides the broader pen strokes for a collected image of renal paracrine signaling. First, we recapitulate the essence of each paracrine system one by one. Thereafter the single components are merged into an overarching physiological concept. The presented survey shows that despite the diversity in the web of paracrine factors, the collected effect on renal function may not be complicated after all. In essence, paracrine activation provides an intelligent system that perceives minor perturbations and reacts with a coordinated and integrated tissue response that relieves the work load from the renal epithelia and favors diuresis and natriuresis. We suggest that the overall function of paracrine signaling is reno-protection and argue that renal paracrine signaling and self-regulation are two sides of the same coin. Thus local paracrine signaling is an intrinsic function of the kidney, and the overall renal effect of changes in blood pressure, volume load, and systemic hormones will always be tinted by its paracrine status.
Collapse
Affiliation(s)
- Jens Leipziger
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| | - Helle Praetorius
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| |
Collapse
|
13
|
Kinguchi S, Wakui H, Azushima K, Haruhara K, Koguchi T, Ohki K, Uneda K, Matsuda M, Haku S, Yamaji T, Yamada T, Kobayashi R, Minegishi S, Ishigami T, Yamashita A, Fujikawa T, Tamura K. Effects of ATRAP in Renal Proximal Tubules on Angiotensin-Dependent Hypertension. J Am Heart Assoc 2019; 8:e012395. [PMID: 30977419 PMCID: PMC6507205 DOI: 10.1161/jaha.119.012395] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background We have previously shown that ATRAP (angiotensin II receptor-associated protein; Agtrap) interacts with AT1R (angiotensin II type 1 receptor) and promotes constitutive internalization of AT 1R so as to inhibit hyperactivation of its downstream signaling. In response to angiotensin II , systemic ATRAP deficiency exacerbates angiotensin II -mediated hypertension via hyperactivation of renal tubular AT 1R. Although ATRAP expression is abundant in renal proximal tubules, little is known about the actual function of renal proximal tubule ATRAP in angiotensin-mediated hypertension. Methods and Results In this study, we examined the in vivo functional role of renal proximal tubule ATRAP in angiotensin-dependent hypertension. We succeeded in generating proximal tubule-specific ATRAP knockout ( PT - KO ) mice for the first time using the Cre/loxP system with Pepck-Cre. Detailed analysis of renal ATRAP expression in PT - KO mice estimated by immunohistochemical and laser-capture microdissection analysis revealed that ATRAP mRNA expression decreased by ≈80% in proximal regions of the nephron in PT - KO mice compared with wild-type ( WT ) mice. We compared blood pressure of PT - KO and WT mice using both tail-cuff and radiotelemetric methods. Blood pressure of PT - KO mice was comparable with that of WT mice at baseline. Moreover, no significant differences were noted in pressor response to angiotensin II (600 ng/kg per min or 1000 ng/kg per minute) infusion between PT - KO and WT mice. In addition, angiotensin II -mediated cardiac hypertrophy was identical between PT - KO and WT mice. Conclusions ATRAP deficiency in proximal tubules did not exacerbate angiotensin-dependent hypertension in vivo. The results indicate that renal proximal tubule ATRAP has a minor role in angiotensin-dependent hypertension in vivo.
Collapse
Affiliation(s)
- Sho Kinguchi
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Hiromichi Wakui
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Kengo Azushima
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan.,2 Cardiovascular and Metabolic Disorders Program Duke-NUS Medical School Singapore Singapore
| | - Kotaro Haruhara
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Tomoyuki Koguchi
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Kohji Ohki
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Kazushi Uneda
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Miyuki Matsuda
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Sona Haku
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Takahiro Yamaji
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Takayuki Yamada
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Ryu Kobayashi
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Shintaro Minegishi
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Tomoaki Ishigami
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Akio Yamashita
- 3 Department of Molecular Biology Yokohama City University Graduate School of Medicine Yokohama Japan
| | - Tetsuya Fujikawa
- 4 Center for Health Service Sciences Yokohama National University Yokohama Japan
| | - Kouichi Tamura
- 1 Department of Medical Science and Cardiorenal Medicine Yokohama City University Graduate School of Medicine Yokohama Japan
| |
Collapse
|
14
|
A new common functional coding variant at the DDC gene change renal enzyme activity and modify renal dopamine function. Sci Rep 2019; 9:5055. [PMID: 30911067 PMCID: PMC6433864 DOI: 10.1038/s41598-019-41504-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 02/05/2019] [Indexed: 01/11/2023] Open
Abstract
The intra-renal dopamine (DA) system is highly expressed in the proximal tubule and contributes to Na+ and blood pressure homeostasis, as well as to the development of nephropathy. In the kidney, the enzyme DOPA Decarboxylase (DDC) originating from the circulation. We used a twin/family study design, followed by polymorphism association analysis at DDC locus to elucidate heritable influences on renal DA production. Dense single nucleotide polymorphism (SNP) genotyping across the DDC locus on chromosome 7p12 was analyzed by re-sequencing guided by trait-associated genetic markers to discover the responsible genetic variation. We also characterized kinetics of the expressed DDC mutant enzyme. Systematic polymorphism screening across the 15-Exon DDC locus revealed a single coding variant in Exon-14 that was associated with DA excretion and multiple other renal traits indicating pleiotropy. When expressed and characterized in eukaryotic cells, the 462Gln variant displayed lower Vmax (maximal rate of product formation by an enzyme) (21.3 versus 44.9 nmol/min/mg) and lower Km (substrate concentration at which half-maximal product formation is achieved by an enzyme.)(36.2 versus 46.8 μM) than the wild-type (Arg462) allele. The highly heritable DA excretion trait is substantially influenced by a previously uncharacterized common coding variant (Arg462Gln) at the DDC gene that affects multiple renal tubular and glomerular traits, and predicts accelerated functional decline in chronic kidney disease.
Collapse
|
15
|
Matsuyama T, Ohashi N, Ishigaki S, Isobe S, Tsuji N, Fujikura T, Tsuji T, Kato A, Miyajima H, Yasuda H. The Relationship between the Intrarenal Dopamine System and Intrarenal Renin-angiotensin System Depending on the Renal Function. Intern Med 2018; 57:3241-3247. [PMID: 29984779 PMCID: PMC6287984 DOI: 10.2169/internalmedicine.0994-18] [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] [Indexed: 01/11/2023] Open
Abstract
Objective The mechanisms underlying the intrarenal renin-angiotensin system (RAS) activation depend on the conditions of kidney diseases. In angiotensin II (AngII) infusion models, the circulating AngII is filtered into the renal tubular lumens, activating intrarenal RAS. However, in the chronic kidney disease (CKD) models, plasma angiotensinogen (AGT) is filtered into the tubular lumens because of glomerular injury, activating intrarenal RAS. The intrarenal dopamine system activation reduces intrarenal AGT expression and suppresses the intrarenal RAS activity in AngII infusion models. However, the relationship between the intrarenal dopamine system and intrarenal RAS has not been elucidated. Therefore, this study was conducted to determine that relationship in CKD patients. Methods We recruited 46 CKD patients (age: 51.1±20.0 years; 16 men; causes of CKD: chronic glomerulonephritis, 34; diabetic nephropathy, 2; nephrosclerosis, 4; and others, 6) not undergoing dialysis or taking RAS blockers. The urinary dopamine (U-DOPA) level, an indicator of intrarenal dopamine activity, and the urinary AGT (U-AGT) level, a surrogate marker of intrarenal RAS activity, were measured. Results As the CKD stages progressed, the U-DOPA levels decreased while the U-AGT levels increased. The U-DOPA levels were significantly and negatively correlated with the U-AGT levels but significantly and positively correlated with the estimated glomerular filtration rate (eGFR). A multiple regression analysis revealed that the U-DOPA levels were associated with the U-AGT levels after adjusting for age, sex, body mass index, and blood pressure (β=-0.38, p=0.045). However, no correlation was observed when eGFR was also adjusted (β=-0.17, p=0.29). Conclusion The negative correlation between the intrarenal dopamine system and intrarenal RAS in CKD patients may be affected by the renal function.
Collapse
Affiliation(s)
| | - Naro Ohashi
- Internal Medicine 1, Hamamatsu University School of Medicine, Japan
| | - Sayaka Ishigaki
- Internal Medicine 1, Hamamatsu University School of Medicine, Japan
| | - Shinsuke Isobe
- Internal Medicine 1, Hamamatsu University School of Medicine, Japan
| | - Naoko Tsuji
- Blood Purification Unit, Hamamatsu University School of Medicine, Japan
| | | | - Takayuki Tsuji
- Internal Medicine 1, Hamamatsu University School of Medicine, Japan
| | - Akihiko Kato
- Blood Purification Unit, Hamamatsu University School of Medicine, Japan
| | - Hiroaki Miyajima
- Internal Medicine 1, Hamamatsu University School of Medicine, Japan
| | - Hideo Yasuda
- Internal Medicine 1, Hamamatsu University School of Medicine, Japan
| |
Collapse
|
16
|
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: 2.3] [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
|
17
|
Hao XQ, Huang CF, Liang F, Deng W, Ou YH, Zhang X, Ding L, Wang D, Wang ST. Dopamine Pretreatment Protects Offspring Rats from LPS-Induced Hypertension and Kidney Damage by Inhibiting NLRP3 Activation in Kidney. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2018. [DOI: 10.1134/s1068162018010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Angiotensin II induces calcium-mediated autophagy in podocytes through enhancing reactive oxygen species levels. Chem Biol Interact 2017; 277:110-118. [DOI: 10.1016/j.cbi.2017.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/25/2017] [Accepted: 09/11/2017] [Indexed: 02/07/2023]
|
19
|
Chen L, Chen DQ, Wang M, Liu D, Chen H, Dou F, Vaziri ND, Zhao YY. Role of RAS/Wnt/β-catenin axis activation in the pathogenesis of podocyte injury and tubulo-interstitial nephropathy. Chem Biol Interact 2017; 273:56-72. [PMID: 28578904 DOI: 10.1016/j.cbi.2017.05.025] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/21/2017] [Accepted: 05/31/2017] [Indexed: 01/11/2023]
|
20
|
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: 71] [Impact Index Per Article: 8.9] [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
|
21
|
Wassenberg T, Willemsen M, Dijkman H, Deinum J, Monnens L. Congenital eyelid ptosis, decreased glomerular filtration, and orthostatic hypotension: Answers. Pediatr Nephrol 2017; 32:1171-1174. [PMID: 27858196 PMCID: PMC5440496 DOI: 10.1007/s00467-016-3515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 08/22/2016] [Accepted: 08/22/2016] [Indexed: 11/17/2022]
Affiliation(s)
- Tessa Wassenberg
- Department of Neurology and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, PO Box 9101, 6500 HB, Nijmegen (935), The Netherlands.
| | - Michèl Willemsen
- 0000 0004 0444 9382grid.10417.33Department of Neurology and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen (935), The Netherlands
| | - Henry Dijkman
- 0000 0004 0444 9382grid.10417.33Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jaap Deinum
- 0000 0004 0444 9382grid.10417.33Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo Monnens
- 0000 0004 0444 9382grid.10417.33Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
22
|
McDonough AA. ISN Forefronts Symposium 2015: Maintaining Balance Under Pressure-Hypertension and the Proximal Tubule. Kidney Int Rep 2016; 1:166-176. [PMID: 27840855 PMCID: PMC5102061 DOI: 10.1016/j.ekir.2016.06.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Renal control of effective circulating volume (ECV) is key for circulatory performance. When renal sodium excretion is inadequate, blood pressure rises and serves as a homeostatic signal to drive natriuresis to re-establish ECV. Recognizing that hypertension involves both renal and vascular dysfunction, this report concerns proximal tubule sodium hydrogen exchanger 3 (NHE3) regulation during acute and chronic hypertension. NHE3 is distributed in tall microvilli (MV) in the proximal tubule, where it reabsorbs a significant fraction of the filtered sodium. NHE3 redistributes, in the plane of the MV membrane, between the MV body, where NHE3 is active, and the MV base, where NHE3 is less active. A high-salt diet and acute hypertension both retract NHE3 to the base and reduce proximal tubule sodium reabsorption independent of a change in abundance. The renin angiotensin system provokes NHE3 redistribution independent of blood pressure: The angiotensin-converting enzyme (ACE) inhibitor captopril redistributes NHE3 to the base and subsequent angiotensin II (AngII) infusion returns NHE3 to the body of the MV and restores reabsorption. Chronic AngII infusion presents simultaneous AngII stimulation and hypertension; that is, NHE3 remains in the body of the MV, due to the high local AngII level and inflammation, and exhibits a compensatory decrease in abundance driven by the hypertension. Genetically modified mice with blunted hypertensive responses to chronic AngII infusion (due to lack of the proximal tubule AngII receptors interleukin-17A or interferon-γ expression) exhibit reduced local AngII accumulation and inflammation and larger decreases in NHE3 abundance, which improves the pressure natriuresis response and reduces the need for elevated blood pressure to facilitate circulating volume balance.
Collapse
Affiliation(s)
- Alicia A McDonough
- Department of Cell and Neurobiology, Keck School of Medicine of the University of Southern California
| |
Collapse
|
23
|
Malagrino PA, Venturini G, Yogi PS, Dariolli R, Padilha K, Kiers B, Gois TC, Cardozo KHM, Carvalho VM, Salgueiro JS, Girardi ACC, Titan SMDO, Krieger JE, Pereira AC. Proteome analysis of acute kidney injury - Discovery of new predominantly renal candidates for biomarker of kidney disease. J Proteomics 2016; 151:66-73. [PMID: 27457269 DOI: 10.1016/j.jprot.2016.07.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/12/2016] [Accepted: 07/18/2016] [Indexed: 02/07/2023]
Abstract
The main bottleneck in studies aiming to identify novel biomarkers in acute kidney injury (AKI) has been the identification of markers that are organ and process specific. Here, we have used different tissues from a controlled porcine renal ischemia/reperfusion (I/R) model to identify new, predominantly renal biomarker candidates for kidney disease. Urine and serum samples were analyzed in pre-ischemia, ischemia (60min) and 4, 11 and 16h post-reperfusion, and renal cortex samples after 24h of reperfusion. Peptides were analyzed on the Q-Exactive™. In renal cortex proteome, we observed an increase in the synthesis of proteins in the ischemic kidney compared to the contralateral, highlighted by transcription factors and epithelial adherens junction proteins. Intersecting the set of proteins up- or down-regulated in the ischemic tissue with both serum and urine proteomes, we identified 6 proteins in the serum that may provide a set of targets for kidney injury. Additionally, we identified 49, being 4 predominantly renal, proteins in urine. As prove of concept, we validated one of the identified biomarkers, dipeptidyl peptidase IV, in a set of patients with diabetic nephropathy. In conclusion, we identified 55 systemic proteins, some of them predominantly renal, candidates for biomarkers of renal disease. BIOLOGICAL SIGNIFICANCE The main bottleneck in studies aiming to identify novel biomarkers in acute kidney injury (AKI) has been the identification of markers that are predominantly renal. In fact, putative biomarkers for this condition have also been identified in a number of other clinical scenarios, such as cardiovascular diseases, chronic kidney failure or in patients being treated in intensive care units from a number of conditions. Here we propose a comprehensive, sequential screening procedure able to identify and validate potential biomarkers for kidney disease, using kidney ischemia/reperfusion as a paradigm for a kidney pathological event.
Collapse
Affiliation(s)
- Pamella Araujo Malagrino
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil.
| | - Gabriela Venturini
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Patrícia Schneider Yogi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Rafael Dariolli
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Bianca Kiers
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Tamiris Carneiro Gois
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | | | | | | | - Adriana Castello Costa Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Silvia Maria de Oliveira Titan
- Renal Division, Department of Clinical Medicine, Faculty of Medicine, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Alexandre Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil.
| |
Collapse
|
24
|
Konkalmatt PR, Asico LD, Zhang Y, Yang Y, Drachenberg C, Zheng X, Han F, Jose PA, Armando I. Renal rescue of dopamine D2 receptor function reverses renal injury and high blood pressure. JCI Insight 2016; 1. [PMID: 27358912 DOI: 10.1172/jci.insight.85888] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dopamine D2 receptor (DRD2) deficiency increases renal inflammation and blood pressure in mice. We show here that long-term renal-selective silencing of Drd2 using siRNA increases renal expression of proinflammatory and profibrotic factors and blood pressure in mice. To determine the effects of renal-selective rescue of Drd2 expression in mice, the renal expression of DRD2 was first silenced using siRNA and 14 days later rescued by retrograde renal infusion of adeno-associated virus (AAV) vector with DRD2. Renal Drd2 siRNA treatment decreased the renal expression of DRD2 protein by 55%, and DRD2 AAV treatment increased the renal expression of DRD2 protein by 7.5- to 10-fold. Renal-selective DRD2 rescue reduced the expression of proinflammatory factors and kidney injury, preserved renal function, and normalized systolic and diastolic blood pressure. These results demonstrate that the deleterious effects of renal-selective Drd2 silencing on renal function and blood pressure were rescued by renal-selective overexpression of DRD2. Moreover, the deleterious effects of 45-minute bilateral ischemia/reperfusion on renal function and blood pressure in mice were ameliorated by a renal-selective increase in DRD2 expression by the retrograde ureteral infusion of DRD2 AAV immediately after the induction of ischemia/reperfusion injury. Thus, 14 days after ischemia/reperfusion injury, the renal expression of profibrotic factors, serum creatinine, and blood pressure were lower in mice infused with DRD2 AAV than in those infused with control AAV. These results indicate an important role of renal DRD2 in limiting renal injury and preserving normal renal function and blood pressure.
Collapse
Affiliation(s)
- Prasad R Konkalmatt
- Department of Medicine, The George Washington University, Washington, DC, USA, and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Laureano D Asico
- Department of Medicine, The George Washington University, Washington, DC, USA, and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yanrong Zhang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yu Yang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Cinthia Drachenberg
- Department of Pathology, University of Maryland Medical Center, Baltimore, Maryland, USA
| | - Xiaoxu Zheng
- Department of Medicine, The George Washington University, Washington, DC, USA, and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Fei Han
- Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Pedro A Jose
- Department of Medicine, The George Washington University, Washington, DC, USA, and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Physiology, The George Washington University, Washington, DC, USA, and University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ines Armando
- Department of Medicine, The George Washington University, Washington, DC, USA, and Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW The kidney mediates the excretion or conservation of water and electrolytes in the face of changing fluid and salt intake and losses. To ultrafilter and reabsorb the exact quantities of free water and salts to maintain euvolemia a range of endocrine, paracrine, and hormonal signaling systems have evolved linking the tubules, capillaries, glomeruli, arterioles, and other intrinsic cells of the kidney. Our understanding of these systems remains incomplete. RECENT FINDINGS Recent work has provided new insights into the workings of the communication pathways between tubular segments and the glomeruli and vasculature, with novel therapeutic agents in development. Particular progress has also been made in the visualization of tubuloglomerular feedback. SUMMARY The review summarizes our current understanding of pathway functions in health and disease, as well as future therapeutic options to protect the healthy and injured kidney.
Collapse
Affiliation(s)
- David A. Ferenbach
- Department of Medicine, Renal Division and Biomedical Engineering Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Joseph V. Bonventre
- Department of Medicine, Renal Division and Biomedical Engineering Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard-Massachusetts Institute of Technology, Division of Health Sciences and Technology, Cambridge, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
26
|
Zhang Y, Jiang X, Qin C, Cuevas S, Jose PA, Armando I. Dopamine D2 receptors' effects on renal inflammation are mediated by regulation of PP2A function. Am J Physiol Renal Physiol 2016; 310:F128-34. [PMID: 26290374 PMCID: PMC4719046 DOI: 10.1152/ajprenal.00453.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 06/15/2015] [Indexed: 01/11/2023] Open
Abstract
Lack or downregulation of the dopamine D2 receptor (D2R) results in increased renal expression of injury markers and proinflammatory factors that is independent of a blood pressure increase. This study aimed to determine the mechanisms involved in the regulation of renal inflammation by D2Rs. Silencing D2Rs in mouse renal proximal tubule cells increased the expression of the proinflammatory TNF-α, monocyte chemoattractant protein-1 (MCP-1), and IL-6. D2R downregulation also increased Akt phosphorylation and activity, and glycogen synthase kinase-3β (GSK3β) phosphorylation and cyclin D1 expression, downstream targets of Akt; however. phosphatidylinositol 3-kinase (PI3K) activity was not affected. Conversely, D2R stimulation decreased Akt and GSK3β phosphorylation and cyclin D1 expression. Increased phospho-Akt, in the absence of increased PI3K activity, may result from decreased Akt dephosphorylation. Inhibition of protein phosphatase 2A (PP2A) with okadaic acid reproduced the effects of D2R downregulation on Akt, GSK3β, and cyclin D1. The PP2A catalytic subunit and regulatory subunit PPP2R2C coimmunoprecipitated with the D2R. Basal phosphatase activity and the expression of PPP2R2C were decreased by D2R silencing that also blunted the increase in phosphatase activity induced by D2R stimulation. Similarly, silencing PPP2R2C also increased the phosphorylation of Akt and GSK3β. Moreover, downregulation of PPP2R2C resulted in increased expression of TNF-α, MCP-1, and IL-6, indicating that decreased phosphatase activity may be responsible for the D2R effect on inflammatory factors. Indeed, the increase in NF-κB reporter activity induced by D2R silencing was blunted by increasing PP2A activity with protamine. Our results show that D2R controls renal inflammation, at least in part, by modulation of the Akt pathway through effects on PP2A activity/expression.
Collapse
Affiliation(s)
- Yanrong Zhang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China; and
| | - Xiaoliang Jiang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China; and
| | - Chuan Qin
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) and Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, P. R. China; and
| | - Santiago Cuevas
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, 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
| | - Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
27
|
Cheng J, Li H, Jie S. Association of the Serum Angiotensin II Level with Disease Severity in Severe Fever with Thrombocytopenia Syndrome Patients. Intern Med 2016; 55:895-900. [PMID: 27086801 DOI: 10.2169/internalmedicine.55.5296] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE Severe fever with thrombocytopenia syndrome (SFTS) is an emerging infectious disease caused by a novel Bunyavirus. Recent data suggest that the physiological balance of multiple proinflammatory cytokines is substantially changed in cases of severe fever with thrombocytopenia syndrome virus (SFTSV) infection, and the inflammatory response probably plays an important role in disease progression. Angiotensin II is an important active substance of the renin-angiotensin system, and studies have demonstrated that angiotensin II is involved in key events in the inflammatory process and can regulate inflammatory cell responses. METHODS In order to elucidate the role of angiotensin II in the pathogenesis of SFTS, we collected serum samples from SFTS patients in the acute or convalescent phase and tested the angiotensin II levels using an enzyme-linked immunosorbent assay as well as SFTSV viral RNA with real-time reverse-transcriptase polymerase chain reaction. Furthermore, we explored possible correlations between the angiotensin II levels and clinical parameters in SFTS patients. RESULTS Our data showed that the serum level of angiotensin II was significantly increased in the acute phase compared with that seen in the convalescent phase and the healthy controls, while there were no significant differences between the convalescent cases and healthy controls (p>0.05). A correlation analysis demonstrated that the level of angiotensin II positively correlated with the SFTS viral RNA load. The angiotensin II levels were also found to be correlated with clinical parameters indicating impairments in organ functions. Moreover, we also found that the angiotensin II levels were significantly increased in the severe cases versus the non-severe cases (p<0.001). CONCLUSION The serum angiotensin II levels in SFTS patients may be used to stratify the disease severity and are possibly predictive of disease outcomes.
Collapse
Affiliation(s)
- Jiamei Cheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | | | | |
Collapse
|
28
|
Muckova P, Wendler S, Rubel D, Büchler R, Alert M, Gross O, Rhode H. Preclinical Alterations in the Serum of COL(IV)A3–/– Mice as Early Biomarkers of Alport Syndrome. J Proteome Res 2015; 14:5202-14. [DOI: 10.1021/acs.jproteome.5b00814] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Petra Muckova
- Institute
of Biochemistry I, University Hospital Jena, Nonnenplan 2-4, 07740 Jena, Germany
- Clinic
of Neurology, University Hospital Jena, Erlanger Allee 101, 07740 Jena, Germany
| | - Sindy Wendler
- Institute
of Biochemistry I, University Hospital Jena, Nonnenplan 2-4, 07740 Jena, Germany
| | - Diana Rubel
- Department
of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Rita Büchler
- Institute
of Biochemistry I, University Hospital Jena, Nonnenplan 2-4, 07740 Jena, Germany
| | - Mandy Alert
- Institute
of Biochemistry I, University Hospital Jena, Nonnenplan 2-4, 07740 Jena, Germany
| | - Oliver Gross
- Department
of Nephrology and Rheumatology, University Medicine Göttingen, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Heidrun Rhode
- Institute
of Biochemistry I, University Hospital Jena, Nonnenplan 2-4, 07740 Jena, Germany
| |
Collapse
|
29
|
Liu X, Wang W, Chen W, Jiang X, Zhang Y, Wang Z, Yang J, Jones JE, Jose PA, Yang Z. Regulation of blood pressure, oxidative stress and AT1R by high salt diet in mutant human dopamine D5 receptor transgenic mice. Hypertens Res 2015; 38:394-9. [PMID: 25716648 PMCID: PMC6400478 DOI: 10.1038/hr.2015.17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/12/2015] [Accepted: 01/25/2015] [Indexed: 12/20/2022]
Abstract
Humans have dopamine D5 receptors (hD5R) with single-nucleotide polymorphisms and a diminished function. We generated hD5(F173L) cDNA that has a decreased response to D5R agonist-mediated increase in cAMP production and increased production of reactive oxygen species, relative to wild-type hD5R (hD5(WT)) cDNA expressed in Chinese hamster ovary cells. To investigate the role of hD5(F173L) in the pathogenesis of salt-sensitive hypertension, we generated transgenic mice overexpressing hD5(F173L) or hD5(WT) and fed them normal (0.8% NaCl) or high (4% NaCl) salt diet. On normal salt diet, the blood pressure, and renal NADPH oxidase activity and angiotensin type 1 receptor (AT1R) expression were higher in hD5(F173L) than hD5(WT) transgenic mice. After 2 weeks on high salt diet, the blood pressure and renal NADPH oxidase activity, but not AT1R expression, were increased in hD5(F173L) but not in hD5(WT) transgenic mice. Candesartan, an AT1R antagonist, decreased the blood pressure and NADPH oxidase activity in hD5(F173L) but not in hD5(WT) transgenic mice. We suggest that the ability of the hD5R to negatively regulate the renal NADPH oxidase activity and AT1R function may have important implications in the pathogenesis of salt-sensitive blood pressure. However, the mechanisms involved in regulating the balance of renal D5R and AT1R function in the oxidative stress-mediated salt-sensitive blood pressure remain to be determined.
Collapse
Affiliation(s)
- Xing Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Wenjie Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Wei Chen
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Xiaoliang Jiang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Yanrong Zhang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Zihao Wang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| | - Jian Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - John E Jones
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pedro A Jose
- 1] Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA [2] Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical Collage (PUMC), Beijing, People's Republic China
| |
Collapse
|
30
|
Choi MR, Kouyoumdzian NM, Rukavina Mikusic NL, Kravetz MC, Rosón MI, Rodríguez Fermepin M, Fernández BE. Renal dopaminergic system: Pathophysiological implications and clinical perspectives. World J Nephrol 2015; 4:196-212. [PMID: 25949933 PMCID: PMC4419129 DOI: 10.5527/wjn.v4.i2.196] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 01/22/2015] [Accepted: 02/04/2015] [Indexed: 02/06/2023] Open
Abstract
Fluid homeostasis, blood pressure and redox balance in the kidney are regulated by an intricate interaction between local and systemic anti-natriuretic and natriuretic systems. Intrarenal dopamine plays a central role on this interactive network. By activating specific receptors, dopamine promotes sodium excretion and stimulates anti-oxidant and anti-inflammatory pathways. Different pathological scenarios where renal sodium excretion is dysregulated, as in nephrotic syndrome, hypertension and renal inflammation, can be associated with impaired action of renal dopamine including alteration in biosynthesis, dopamine receptor expression and signal transduction. Given its properties on the regulation of renal blood flow and sodium excretion, exogenous dopamine has been postulated as a potential therapeutic strategy to prevent renal failure in critically ill patients. The aim of this review is to update and discuss on the most recent findings about renal dopaminergic system and its role in several diseases involving the kidneys and the potential use of dopamine as a nephroprotective agent.
Collapse
|
31
|
Yao M, Wang X, Wang X, Zhang T, Chi Y, Gao F. The Notch pathway mediates the angiotensin II-induced synthesis of extracellular matrix components in podocytes. Int J Mol Med 2015; 36:294-300. [PMID: 25902289 DOI: 10.3892/ijmm.2015.2193] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 04/15/2015] [Indexed: 11/06/2022] Open
Abstract
The Notch pathway is known to contribute to the development of glomerular disease. Angiotensin II (Ang II), an important member of the renin-angiotensin system, stimulates the accumulation of extracellular matrix components in glomerular disease; however, the exact mechanisms involved remain to be elucidated. In the present study, we aimed to investigate the effects of the Notch pathway on the synthesis of extracellular matrix components in Ang II-stimulated podocytes. Mouse podocytes were stimulated with Ang II (10-6 mol/l). The activation of the Notch pathway was inhibited by a vector carrying short hairpin RNA (shRNA) targeting Notch1 (sh-Notch1) or by γ-secretase inhibitor (GSI). The protein levels of Notch1, Notch intracellular domain 1 (NICD1), hairy and enhancer of split-1 (Hes1), matrix metalloproteinase (MMP)-2, MMP-9, transforming growth factor-β1 (TGF-β1), type IV collagen and laminin were determined by western blot analysis. The Notch1, Hes1, MMP-2, MMP-9, TGF-β1, type IV collagen and laminin mRNA levels were detected by RT-PCR. The MMP-2 and MMP-9 activity was measured using a cell active fluorescence assay kit. The levels of TGF-β1, type IV collagen and laminin were determined in the culture medium of the podocytes by enzyme-linked immunosorbent assay (ELISA). Our results revealed that Ang II upregulated Notch1, NICD1, Hes1, TGF-β1, type IV collagen and laminin expression and downregulated MMP-2 and MMP-9 expression in the cultured podocytes. The inhibition of the Notch pathway by sh-Notch1 or GSI increased MMP-2 and MMP-9 expression, decreased the TGF-β1 level and suppressed type IV collagen and laminin expression. The inhibition of the Notch pathway by sh-Notch1 or GSI also increased MMP-2 and MMP-9 activity, and decreased TGF-β1 levels, type IV collagen levels and laminin secretion. These findings indicate that the Notch pathway potentially mediates the Ang II-induced synthesis of extracellular matrix components in podocytes through the regulation of MMPs and TGF-β1.
Collapse
Affiliation(s)
- Min Yao
- Department of Pathology, Τhe Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Xiaomei Wang
- Department of Pathology, Τhe Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Xiaomeng Wang
- Department of Pathology, Τhe Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Tao Zhang
- Department of Nephrology, Τhe Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yanqing Chi
- Department of Nephrology, Τhe Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Feng Gao
- Department of Pathology, Τhe Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
32
|
Han F, Konkalmatt P, Chen J, Gildea J, Felder RA, Jose PA, Armando I. MiR-217 mediates the protective effects of the dopamine D2 receptor on fibrosis in human renal proximal tubule cells. Hypertension 2015; 65:1118-25. [PMID: 25801876 DOI: 10.1161/hypertensionaha.114.05096] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/01/2015] [Indexed: 01/11/2023]
Abstract
Lack or downregulation of the dopamine D2 receptor (D2R) increases the vulnerability to renal inflammation independent of blood pressure in mice. Common single nucleotide polymorphisms (SNPs) rs6276, 6277, and 1800497 in the human D2R gene are associated with decreased receptor expression/function and hypertension. Human renal proximal tubule cells from subjects carrying these SNPs have decreased D2R expression and increased expression of profibrotic factors and production of extracellular matrix proteins. We tested the hypothesis that the D2R mediates these effects by regulating micro-RNA expression. In cells carrying D2R SNPs, micro-RNAs (miRs)-217, miR-224, miR-335, and miR-1265 were downregulated, whereas miR-1290 was upregulated >4-fold compared with those carrying D2R wild-type alleles. However, only miR-217 was directly regulated by D2R expression. In cells carrying D2R wild-type, miR-217 inhibitor increased the expression of transforming growth factor (TGF)-β1, matrix metalloproteinase 3, fibronectin 1, and collagen 1a, whereas miR-217 mimic had the opposite effect. In cells carrying D2R SNPs, miR-217 mimic also decreased the expression of TGFβ1 and its targets. Wnt5a, a miR-217 target, was increased in cells carrying D2R SNPs and decreased by miR-217 mimic but increased by miR-217 inhibitor in both cell types. In cells carrying D2R wild-type, Wnt5a treatment increased TGFβ1 while silencing Ror2, a Wnt5a receptor, decreased TGFβ1 and blunted the Wnt5a-induced increase in cells carrying D2R wild-type. Our results show that renal proximal tubule cells from subjects carrying D2R SNPs resulting in D2R downregulation have increased TGFβ1 that is mediated by decreased regulation of the miR-217-Wnt5a-Ror2 pathway.
Collapse
Affiliation(s)
- Fei Han
- From the Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (F.H., J.C.); Division of Nephrology, Department of Medicine (P.K., P.A.J., I.A.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore; and Department of Pathology, University of Virginia School of Medicine, Charlottesville (J.G., R.A.F.)
| | - Prasad Konkalmatt
- From the Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (F.H., J.C.); Division of Nephrology, Department of Medicine (P.K., P.A.J., I.A.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore; and Department of Pathology, University of Virginia School of Medicine, Charlottesville (J.G., R.A.F.)
| | - Jianghua Chen
- From the Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (F.H., J.C.); Division of Nephrology, Department of Medicine (P.K., P.A.J., I.A.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore; and Department of Pathology, University of Virginia School of Medicine, Charlottesville (J.G., R.A.F.)
| | - John Gildea
- From the Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (F.H., J.C.); Division of Nephrology, Department of Medicine (P.K., P.A.J., I.A.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore; and Department of Pathology, University of Virginia School of Medicine, Charlottesville (J.G., R.A.F.)
| | - Robin A Felder
- From the Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (F.H., J.C.); Division of Nephrology, Department of Medicine (P.K., P.A.J., I.A.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore; and Department of Pathology, University of Virginia School of Medicine, Charlottesville (J.G., R.A.F.)
| | - Pedro A Jose
- From the Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (F.H., J.C.); Division of Nephrology, Department of Medicine (P.K., P.A.J., I.A.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore; and Department of Pathology, University of Virginia School of Medicine, Charlottesville (J.G., R.A.F.)
| | - Ines Armando
- From the Kidney Disease Center, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China (F.H., J.C.); Division of Nephrology, Department of Medicine (P.K., P.A.J., I.A.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore; and Department of Pathology, University of Virginia School of Medicine, Charlottesville (J.G., R.A.F.)
| |
Collapse
|
33
|
Cuevas CA, Gonzalez AA, Inestrosa NC, Vio CP, Prieto MC. Angiotensin II increases fibronectin and collagen I through the β-catenin-dependent signaling in mouse collecting duct cells. Am J Physiol Renal Physiol 2014; 308:F358-65. [PMID: 25411386 DOI: 10.1152/ajprenal.00429.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The contribution of angiotensin II (ANG II) to renal and tubular fibrosis has been widely reported. Recent studies have shown that collecting duct cells can undergo mesenchymal transition suggesting that collecting duct cells are involved in interstitial fibrosis. The Wnt/β-catenin signaling pathway plays an essential role in development, organogenesis, and tissue homeostasis; however, the dysregulation of this pathway has been linked to fibrosis. In this study, we investigated whether AT1 receptor activation induces the expression of fibronectin and collagen I via the β-catenin pathway in mouse collecting duct cell line M-1. ANG II (10(-7) M) treatment in M-1 cells increased mRNA, protein levels of fibronectin and collagen I, the β-catenin target genes (cyclin D1 and c-myc), and the myofibroblast phenotype. These effects were prevented by candesartan, an AT1 receptor blocker. Inhibition of the β-catenin degradation with pyrvinium pamoate (pyr; 10(-9) M) prevented the ANG II-induced expression of fibronectin, collagen I, and β-catenin target genes. ANG II treatment promoted the accumulation of β-catenin protein in a time-dependent manner. Because phosphorylation of glycogen synthase kinase-3β (GSK-3β) inhibits β-catenin degradation, we further evaluated the effects of ANG II and ANG II plus pyr on p-ser9-GSK-3β levels. ANG II-dependent upregulation of β-catenin protein levels was correlated with GSK-3β phosphorylation. These effects were prevented by pyr. Our data indicate that in M-1 collecting duct cells, the β-catenin pathway mediates the stimulation of fibronectin and collagen I in response to AT1 receptor activation.
Collapse
Affiliation(s)
- Catherina A Cuevas
- Department of Physiology, Center of Aging and Regeneration CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile; Department of Cell and Molecular Biology, Center of Aging and Regeneration CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile; Department of Physiology, Tulane University, New Orleans, Louisiana; and
| | - Alexis A Gonzalez
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Nibaldo C Inestrosa
- Department of Cell and Molecular Biology, Center of Aging and Regeneration CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos P Vio
- Department of Physiology, Center of Aging and Regeneration CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Minolfa C Prieto
- Department of Physiology, Tulane University, New Orleans, Louisiana; and Department of Hypertension and Renal Center of Excellence, Tulane University, New Orleans, Louisiana
| |
Collapse
|
34
|
Rukavina Mikusic NL, Kravetz MC, Kouyoumdzian NM, Della Penna SL, Rosón MI, Fernández BE, Choi MR. 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: 1.1] [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
Affiliation(s)
- N. L. Rukavina Mikusic
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - M. C. Kravetz
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - N. M. Kouyoumdzian
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - S. L. Della Penna
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - M. I. Rosón
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - B. E. Fernández
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| | - M. R. Choi
- Department of Pathophysiology, Faculty of Pharmacy and Biochemistry, University of Buenos Aires, CONICET, INFIBIOC, 1113 Buenos Aires, Argentina
| |
Collapse
|
35
|
Gao N, Wang H, Zhang X, Yang Z. The inhibitory effect of angiotensin II on BKCa channels in podocytes via oxidative stress. Mol Cell Biochem 2014; 398:217-22. [PMID: 25234195 DOI: 10.1007/s11010-014-2221-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 09/13/2014] [Indexed: 01/11/2023]
Abstract
Angiotensin II (Ang II) is an important active substance of the renin-angiotensin system (RAS). The present study has confirmed that abnormalities of Ang II may be related with cerebrovascular diseases, endocrine diseases, cardiovascular diseases, liver diseases, such as: cerebral hypoxia, diabetes, obesity, atrial fibrillation, and liver cirrhosis. However, understanding effects of Ang II on podocytes is not enough. This study was to investigate the effects of oxidative stress on the large conductance, Ca(2+)-activated K(+) channels (BKCa). Results from the 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay showed that Ang II induced podocyte death in a concentration-dependent manner. The measurement of superoxide dismutase (SOD) generation demonstrated that Ang II decreased the total SOD of cellular levels. Meaningfully, pretreatment of a type of ROS scavenger formulations named N-(mercaptopropionyl)-glycine (N-MPG) could inhibit podocyte apoptosis induced by Ang II. Meanwhile, patch-clamp technique was used in this study to detect the effects of Ang II on currents of BKCa channel in podocytes. The results indicated that Ang II inhibited the current amplitude of BKCa channel and decreased the slope of I-V curve. Ang II also made the activation curves of BKCa channel shift to the left. These results may provide a theoretical basis for potential treatment of chronic glomerular disease in the future.
Collapse
Affiliation(s)
- Na Gao
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University, Tianjin, 300071, China
| | | | | | | |
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an update on the current knowledge regarding the role of the intrarenal rennin-angiotensin system (RAS) in the regulation of glomerular function including glomerular dynamics and filtration rate, glomerular permeability and structural alterations during chronic increases in intrarenal angiotensin (Ang) II. RECENT FINDINGS Recent studies have continued to delineate the complex interactions among the various RAS components that participate in regulating glomerular function. Although Ang II acting on AT1 receptors remains as the predominant influence on glomerular dynamics, some of these effects are indirectly mediated by Ang II modulating the sensitivity of the macula densa tubuloglomerular feedback mechanism as well as the more recently described feedback mechanism from the connecting tubule. Interestingly, the actions of Ang II on these systems cause opposite effects on glomerular function demonstrating the complexities associated with the influences of Ang II on glomerular function. When chronically elevated, Ang II also stimulates and/or interacts with other factors, including reactive oxygen species, cytokines and growth factors and other hormones or paracrine agents, to elicit structural alterations. SUMMARY Recent studies have provided further evidence for the presence of many components of the RAS in glomerular structures, which supports the importance of locally produced angiotensin peptides to regulate glomerular haemodynamics, filtration rate and macromolecular permeability and contribute to fibrosis and glomerular injury when inappropriately augmented.
Collapse
|
37
|
Jennings BL, George LW, Pingili AK, Khan NS, Estes AM, Fang XR, Gonzalez FJ, Malik KU. Estrogen metabolism by cytochrome P450 1B1 modulates the hypertensive effect of angiotensin II in female mice. Hypertension 2014; 64:134-40. [PMID: 24777982 DOI: 10.1161/hypertensionaha.114.03275] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To determine the role of cytochrome P450 (CYP) 1B1 in the sex difference in response to angiotensin II (Ang II)-induced hypertension, female Cyp1b1(+/+) and Cyp1b1(-/-) mice were infused with Ang II (700 ng/kg per minute) or vehicle with or without ovariectomy. In addition, mice were treated with the CYP1B1 inhibitor, 2,3',4,5'-tetramethoxystilbene (TMS; 300 μg/kg IP, every third day), and 17-β estradiol metabolites, 2-hydroxyestradiol (2-OHE), 4-OHE, or 2-methoxyestradiol (1.5 mg/kg per day IP, for 2 weeks) and systolic blood pressure (SBP) measured. Ang II increased SBP more in Cyp1b1(-/-) than in Cyp1b1(+/+) mice (119±3-171±11 versus 120±4-149±4 mm Hg; P<0.05). Ang II caused cardiovascular remodeling and endothelial dysfunction and increased vascular reactivity and oxidative stress in Cyp1b1(-/-) but not in Cyp1b1(+/+)mice. The Ang II-induced increase in SBP was enhanced by ovariectomy and TMS in Cyp1b1(+/+) but not in Cyp1b1(-/-) mice. 2-OHE did not alter Ang II-induced increase in SBP in Cyp1b1(+/+) mice but minimized it in Cyp1b1(-/-) mice, whereas 4-OHE enhanced Ang II-induced increase in SBP in Cyp1b1(+/+) mice but did not alter the increased SBP in Cyp1b1(-/-) mice. 2-OHE-derived catechol-O-methyltransferase metabolite, 2-methoxyestradiol, inhibited Ang II-induced increase in SBP in Cyp1b1(-/-) mice. Ang II increased plasma levels of 2-methoxyestradiol in Cyp1b1(+/+) but not in Cyp1b1(-/-) mice and increased activity of cardiac extracellular signal-regulated kinase 1/2, p38 mitogen-activated kinase, c-Src, and Akt in Cyp1b1(-/-) but not in Cyp1b1(+/+) mice. These data suggest that CYP1B1 protects against Ang II-induced hypertension and associated cardiovascular changes in female mice, most likely mediated by 2-methoxyestradiol-inhibiting oxidative stress and the activity of these signaling molecules.
Collapse
Affiliation(s)
- Brett L Jennings
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - L Watson George
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Ajeeth K Pingili
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Nayaab S Khan
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Anne M Estes
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Xiao R Fang
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Frank J Gonzalez
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.)
| | - Kafait U Malik
- From the Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis (B.L.J., L.W.G., A.K.P., N.S.K., A.M.E., X.R.F., K.U.M.); and Laboratory of Metabolism, National Cancer Institute, Bethesda, MD (F.J.G.).
| |
Collapse
|
38
|
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: 6.0] [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
|
39
|
Skov J, Persson F, Frøkiær J, Christiansen JS. Tissue Renin-Angiotensin systems: a unifying hypothesis of metabolic disease. Front Endocrinol (Lausanne) 2014; 5:23. [PMID: 24592256 PMCID: PMC3938116 DOI: 10.3389/fendo.2014.00023] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/13/2014] [Indexed: 01/11/2023] Open
Abstract
The actions of angiotensin peptides are diverse and locally acting tissue renin-angiotensin systems (RAS) are present in almost all tissues of the body. An activated RAS strongly correlates to metabolic disease (e.g., diabetes) and its complications and blockers of RAS have been demonstrated to prevent diabetes in humans. Hyperglycemia, obesity, hypertension, and cortisol are well-known risk factors of metabolic disease and all stimulate tissue RAS whereas glucagon-like peptide-1, vitamin D, and aerobic exercise are inhibitors of tissue RAS and to some extent can prevent metabolic disease. Furthermore, an activated tissue RAS deteriorates the same risk factors creating a system with several positive feedback pathways. The primary effector hormone of the RAS, angiotensin II, stimulates reactive oxygen species, induces tissue damage, and can be associated to most diabetic complications. Based on these observations, we hypothesize that an activated tissue RAS is the principle cause of metabolic syndrome and type 2 diabetes, and additionally is mediating the majority of the metabolic complications. The involvement of positive feedback pathways may create a self-reinforcing state and explain why metabolic disease initiate and progress. The hypothesis plausibly unifies the major predictors of metabolic disease and places tissue RAS regulation in the center of metabolic control.
Collapse
Affiliation(s)
- Jeppe Skov
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
- Novo Nordisk A/S, Bagsvaerd, Denmark
- *Correspondence: Jeppe Skov, Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Norrebrogade 44, Aarhus DK-8000, Denmark e-mail:
| | | | - Jørgen Frøkiær
- Department of Clinical Physiology and Molecular Imaging, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | |
Collapse
|
40
|
Jiang X, Konkalmatt P, Yang Y, Gildea J, Jones JE, Cuevas S, Felder RA, Jose PA, Armando I. Single-nucleotide polymorphisms of the dopamine D2 receptor increase inflammation and fibrosis in human renal proximal tubule cells. Hypertension 2013; 63:e74-80. [PMID: 24379187 DOI: 10.1161/hypertensionaha.113.02569] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The dopamine D2 receptor (D2R) negatively regulates inflammation in mouse renal proximal tubule cells (RPTCs), and lack or downregulation of the receptor in mice increases the vulnerability to renal inflammation independent of blood pressure. Some common single-nucleotide polymorphisms (SNPs; rs6276, rs6277, and rs1800497) in the human DRD2 gene are associated with decreased D2R expression and function, as well as high blood pressure. We tested the hypothesis that human RPTCs (hRPTCs) expressing these SNPs have increased expression of inflammatory and injury markers. We studied immortalized hRPTCs carrying D2R SNPs and compared them with cells carrying no D2R SNPs. RPTCs with D2R SNPs had decreased D2R expression and function. The expressions of the proinflammatory tumor necrosis factor-α and the profibrotic transforming growth factor-β1 and its signaling targets Smad3 and Snail1 were increased in hRPTC with D2R SNPs. These cells also showed induction of epithelial mesenchymal transition and production of extracellular matrix proteins, assessed by increased vimentin, fibronectin 1, and collagen I a1. To test the specificity of these D2R SNP effects, hRPTC with D2R SNPs were transfected with a plasmid encoding wild-type DRD2. The expression of D2R was increased and that of transforming growth factor-β1, Smad3, Snail1, vimentin, fibronectin 1, and collagen I a1 was decreased in hRPTC with D2R SNPs transfected with wild-type DRD2 compared with hRPTC-D2R SNP transfected with empty vector. These data support the hypothesis that D2R function has protective effects in hRPTCs and suggest that carriers of these SNPs may be prone to chronic renal disease and high blood pressure.
Collapse
Affiliation(s)
- Xiaoliang Jiang
- University of Maryland School of Medicine, Department of Medicine, Division of Nephrology, 20 Penn St- HSFII Suite S003C, Baltimore, MD 21201.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
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: 5.7] [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
|
42
|
Aromatic L-amino acid decarboxylase (AADC) is crucial for brain development and motor functions. PLoS One 2013; 8:e71741. [PMID: 23940784 PMCID: PMC3734303 DOI: 10.1371/journal.pone.0071741] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/02/2013] [Indexed: 01/01/2023] Open
Abstract
Aromatic L-amino acid decarboxylase (AADC) deficiency is a rare pediatric neuro-metabolic disease in children. Due to the lack of an animal model, its pathogenetic mechanism is poorly understood. To study the role of AADC in brain development, a zebrafish model of AADC deficiency was generated. We identified an aadc gene homolog, dopa decarboxylase (ddc), in the zebrafish genome. Whole-mount in situ hybridization analysis showed that the ddc gene is expressed in the epiphysis, locus caeruleus, diencephalic catecholaminergic clusters, and raphe nuclei of 36-h post-fertilization (hpf) zebrafish embryos. Inhibition of Ddc by AADC inhibitor NSD-1015 or anti-sense morpholino oligonucleotides (MO) reduced brain volume and body length. We observed increased brain cell apoptosis and loss of dipencephalic catecholaminergic cluster neurons in ddc morphants (ddc MO-injected embryos). Seizure-like activity was also detected in ddc morphants in a dose-dependent manner. ddc morphants had less sensitive touch response and impaired swimming activity that could be rescued by injection of ddc plasmids. In addition, eye movement was also significantly impaired in ddc morphants. Collectively, loss of Ddc appears to result in similar phenotypes as that of ADCC deficiency, thus zebrafish could be a good model for investigating pathogenetic mechanisms of AADC deficiency in children.
Collapse
|
43
|
Zhang BQ, Wang G, Zhang JP, Hu JY, Xiao R, Lei ZY, Ruan J, Dang YM, Zhang DX, Bian XW, Huang YS. Protective effects of enalapril, an angiotensin-converting enzyme inhibitor, on multiple organ damage following scald injury in rats. Biotechnol Appl Biochem 2013; 59:307-13. [PMID: 23586864 DOI: 10.1002/bab.1027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2012] [Accepted: 06/14/2012] [Indexed: 01/11/2023]
Abstract
The aim of this study is to investigate the effects of enalapril, an angiotensin-converting enzyme inhibitor, on multiple organ damage after scald injury. Healthy adult rats (half male and half female; 8-12 weeks old) were randomly assigned to the following treatments: sham operation, scald injury, and intraperitoneal enalapril (1, 2, and 4 mg/kg body weight) treatment after scalding. At 1, 12, and 24 H postscald, left ventricular and aortic hemodynamics were measured using a multichannel physiological recorder. Functional and pathological changes of the heart, liver, and kidney were examined by biochemical and histological methods. Compared with sham controls, untreated scalded animals showed decreased hemodynamic parameters and increased myocardial angiotensin II, serum creatine kinase heart isoenzyme, and serum cardiac troponin I and histopathological inflammation in the myocardium 12 H postscald. These hemodynamic, functional, and pathological changes were attenuated by 1 mg/kg enalapril. Enalapril reversed scald-induced elevations in aspartate aminotransferase, alanine aminotransferase, blood urea nitrogen, and blood creatinine 12 H postscald, and ameliorated focal necrosis in the liver and erythrocyte cast formation in renal tubules. However, higher doses of enalapril yielded less or no improvement in organ dysfunction. Enalapril at 1 mg/kg attenuates scald-induced multiple organ damage in rats.
Collapse
Affiliation(s)
- Bing-Qian Zhang
- Institute of Burn Research of PLA, National Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Campanholle G, Ligresti G, Gharib SA, Duffield JS. Cellular mechanisms of tissue fibrosis. 3. Novel mechanisms of kidney fibrosis. Am J Physiol Cell Physiol 2013; 304:C591-603. [PMID: 23325411 DOI: 10.1152/ajpcell.00414.2012] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease, defined as loss of kidney function for more than three months, is characterized pathologically by glomerulosclerosis, interstitial fibrosis, tubular atrophy, peritubular capillary rarefaction, and inflammation. Recent studies have identified a previously poorly appreciated, yet extensive population of mesenchymal cells, called either pericytes when attached to peritubular capillaries or resident fibroblasts when embedded in matrix, as the progenitors of scar-forming cells known as myofibroblasts. In response to sustained kidney injury, pericytes detach from the vasculature and differentiate into myofibroblasts, a process not only causing fibrosis, but also directly contributing to capillary rarefaction and inflammation. The interrelationship of these three detrimental processes makes myofibroblasts and their pericyte progenitors an attractive target in chronic kidney disease. In this review, we describe current understanding of the mechanisms of pericyte-to-myofibroblast differentiation during chronic kidney disease, draw parallels with disease processes in the glomerulus, and highlight promising new therapeutic strategies that target pericytes or myofibroblasts. In addition, we describe the critical paracrine roles of epithelial, endothelial, and innate immune cells in the fibrogenic process.
Collapse
Affiliation(s)
- Gabriela Campanholle
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | | | | |
Collapse
|
45
|
Arora MK, Singh UK. Molecular mechanisms in the pathogenesis of diabetic nephropathy: an update. Vascul Pharmacol 2013; 58:259-71. [PMID: 23313806 DOI: 10.1016/j.vph.2013.01.001] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 01/04/2013] [Accepted: 01/04/2013] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus is known to trigger retinopathy, neuropathy and nephropathy. Diabetic nephropathy, a long-term major microvascular complication of uncontrolled hyperglycemia, affects a large population worldwide. Recent findings suggest that numerous pathways are activated during the course of diabetes mellitus and that these pathways individually or collectively play a role in the induction and progression of diabetic nephropathy. However, clinical strategies targeting these pathways to manage diabetic nephropathy remain unsatisfactory, as the number of diabetic patients with nephropathy is increasing yearly. To develop ground-breaking therapeutic options to prevent the development and progression of diabetic nephropathy, a comprehensive understanding of the molecular mechanisms involved in the pathogenesis of the disease is mandatory. Therefore, the purpose of this paper is to discuss the underlying mechanisms and downstream pathways involved in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Mandeep Kumar Arora
- Faculty of Pharmacy, Swami Vivekanand Subharti University, Meerut 250005, Uttar Pradesh, India.
| | | |
Collapse
|
46
|
Duffield JS, Lupher M, Thannickal VJ, Wynn TA. Host responses in tissue repair and fibrosis. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:241-76. [PMID: 23092186 DOI: 10.1146/annurev-pathol-020712-163930] [Citation(s) in RCA: 463] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Myofibroblasts accumulate in the spaces between organ structures and produce extracellular matrix (ECM) proteins, including collagen I. They are the primary "effector" cells in tissue remodeling and fibrosis. Previously, leukocyte progenitors termed fibrocytes and myofibroblasts generated from epithelial cells through epithelial-to-mesenchymal transition (EMT) were considered the primary sources of ECM-producing myofibroblasts in injured tissues. However, genetic fate mapping experiments suggest that mesenchyme-derived cells, known as resident fibroblasts, and pericytes are the primary precursors of scar-forming myofibroblasts, whereas epithelial cells, endothelial cells, and myeloid leukocytes contribute to fibrogenesis predominantly by producing key fibrogenic cytokines and by promoting cell-to-cell communication. Numerous cytokines derived from T cells, macrophages, and other myeloid cell populations are important drivers of myofibroblast differentiation. Monocyte-derived cell populations are key regulators of the fibrotic process: They act as a brake on the processes driving fibrogenesis, and they dismantle and degrade established fibrosis. We discuss the origins, modes of activation, and fate of myofibroblasts in various important fibrotic diseases and describe how manipulation of macrophage activation could help ameliorate fibrosis.
Collapse
Affiliation(s)
- Jeremy S Duffield
- Division of Nephrology, Center for Lung Biology, and the Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98019, USA
| | | | | | | |
Collapse
|
47
|
Zhang Y, Cuevas S, Asico LD, Escano C, Yang Y, Pascua AM, Wang X, Jones JE, Grandy D, Eisner G, Jose PA, Armando I. Deficient dopamine D2 receptor function causes renal inflammation independently of high blood pressure. PLoS One 2012; 7:e38745. [PMID: 22719934 PMCID: PMC3375266 DOI: 10.1371/journal.pone.0038745] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 05/10/2012] [Indexed: 12/15/2022] Open
Abstract
Renal dopamine receptors participate in the regulation of blood pressure. Genetic factors, including polymorphisms of the dopamine D(2) receptor gene (DRD2) are associated with essential hypertension, but the mechanisms of their contribution are incompletely understood. Mice lacking Drd2 (D(2)-/-) have elevated blood pressure, increased renal expression of inflammatory factors, and renal injury. We tested the hypothesis that decreased dopamine D(2) receptor (D(2)R) function increases vulnerability to renal inflammation independently of blood pressure, is an immediate cause of renal injury, and contributes to the subsequent development of hypertension. In D(2)-/- mice, treatment with apocynin normalized blood pressure and decreased oxidative stress, but did not affect the expression of inflammatory factors. In mouse RPTCs Drd2 silencing increased the expression of TNFα and MCP-1, while treatment with a D(2)R agonist abolished the angiotensin II-induced increase in TNF-α and MCP-1. In uni-nephrectomized wild-type mice, selective Drd2 silencing by subcapsular infusion of Drd2 siRNA into the remaining kidney produced the same increase in renal cytokines/chemokines that occurs after Drd2 deletion, increased the expression of markers of renal injury, and increased blood pressure. Moreover, in mice with two intact kidneys, short-term Drd2 silencing in one kidney, leaving the other kidney undisturbed, induced inflammatory factors and markers of renal injury in the treated kidney without increasing blood pressure. Our results demonstrate that the impact of decreased D(2)R function on renal inflammation is a primary effect, not necessarily associated with enhanced oxidant activity, or blood pressure; renal damage is the cause, not the result, of hypertension. Deficient renal D(2)R function may be of clinical relevance since common polymorphisms of the human DRD2 gene result in decreased D(2)R expression and function.
Collapse
Affiliation(s)
- Yanrong Zhang
- Division of Nephrology, Department of Medicine, School of Medicine, University of Maryland, Baltimore, Maryland, United States of America
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|