1
|
Sun X, Wu C, Tian X, Wang P, Guo J, Shao Z, Wei Q. Activation of Dopamine Receptor D1 and Downstream Cellular Functions by Polydopamine. ACS Biomater Sci Eng 2024; 10:420-428. [PMID: 38142403 DOI: 10.1021/acsbiomaterials.3c01615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2023]
Abstract
Polydopamine is a remarkable molecule that has gained considerable attention for its role in material surface modification, leading to an abundance of research in the biomaterial domain. While its widespread use is well documented, the molecule's potential cellular interactions have been less explored. In particular, dopamine serves as a neurotransmitter and a hormone that interacts with dopamine receptors in cells. Our study sheds light on the previously unexamined interaction between polydopamine and dopamine receptor D1 (DRD1). We discovered that polydopamine, along with its derivatives, such as levodopa and catechol, can activate DRD1─a function previously attributed solely to dopamine. Moreover, we found that polydopamine has the ability to influence cell behavior through the cAMP/PKA pathway, thereby affecting RhoA activity and stress fiber formation. These observations invite further consideration regarding the biological safety of polydopamine in biomedical contexts and also open avenues for new research directions in designing bioactive functional materials.
Collapse
Affiliation(s)
- Xin Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Chao Wu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Junling Guo
- College of Biomass Science and Engineering, Sichuan University, Chengdu, Sichuan 610065, China
- Department of Chemical and Biological Engineering, University of British, Columbia Vancouver, BC V6T 1Z4, Canada
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
2
|
Rasmi Y, Shokati A, Hatamkhani S, Farnamian Y, Naderi R, Jalali L. Assessment of the relationship between the dopaminergic pathway and severe acute respiratory syndrome coronavirus 2 infection, with related neuropathological features, and potential therapeutic approaches in COVID-19 infection. Rev Med Virol 2024; 34:e2506. [PMID: 38282395 DOI: 10.1002/rmv.2506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 07/06/2023] [Accepted: 12/17/2023] [Indexed: 01/30/2024]
Abstract
Dopamine is a known catecholamine neurotransmitter involved in several physiological processes, including motor control, motivation, reward, cognition, and immune function. Dopamine receptors are widely distributed throughout the nervous system and in immune cells. Several viruses, including human immunodeficiency virus and Japanese encephalitis virus, can use dopaminergic receptors to replicate in the nervous system and are involved in viral neuropathogenesis. In addition, studies suggest that dopaminergic receptors may play a role in the progression and pathogenesis of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. When SARS-CoV-2 binds to angiotensin-converting enzyme 2 receptors on the surface of neuronal cells, the spike protein of the virus can bind to dopaminergic receptors on neighbouring cells to accelerate its life cycle and exacerbate neurological symptoms. In addition, recent research has shown that dopamine is an important regulator of the immune-neuroendocrine system. Most immune cells express dopamine receptors and other dopamine-related proteins, indicating the importance of dopaminergic immune regulation. The increase in dopamine concentration during SARS-CoV2 infection may reduce immunity (innate and adaptive) that promotes viral spread, which could lead to neuronal damage. In addition, dopaminergic signalling in the nervous system may be affected by SARS-CoV-2 infection. COVID -19 can cause various neurological symptoms as it interacts with the immune system. One possible treatment strategy for COVID -19 patients could be the use of dopamine antagonists. To fully understand how to protect the neurological system and immune cells from the virus, we need to study the pathophysiology of the dopamine system in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yousef Rasmi
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ameneh Shokati
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shima Hatamkhani
- Experimental and Applied Pharmaceutical Sciences Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Yeganeh Farnamian
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Roya Naderi
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ladan Jalali
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
3
|
Mood and behavior regulation: interaction of lithium and dopaminergic system. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023:10.1007/s00210-023-02437-1. [PMID: 36843130 DOI: 10.1007/s00210-023-02437-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/20/2023] [Indexed: 02/28/2023]
Abstract
Lithium is one of the most effect mood-stabilizing drugs prescribed especially for bipolar disorder. Lithium has wide range effects on different molecular factors and neural transmission including dopaminergic signaling. On the other hand, mesolimbic and mesocortical dopaminergic signaling is significantly involved in the pathophysiology of neuropsychiatric disorders. This review article aims to study lithium therapeutic mechanisms, dopaminergic signaling, and the interaction of lithium and dopamine. We concluded that acute and chronic lithium treatments often reduce dopamine synthesis and level in the brain. However, some studies have reported conflicting results following lithium treatment, especially chronic treatment. The dosage, duration, and type of lithium administration, and the brain region selected for measuring dopamine level were not significant differences in different chronic treatments used in previous studies. It was suggested that lithium has various mechanisms affecting dopaminergic signaling and mood, and that many molecular factors can be involved, including brain-derived neurotrophic factor (BDNF), cAMP response element-binding protein (CREB), β-catenin, protein kinase B (Akt), and glycogen synthase kinase-3 beta (GSK-3β). Thus, molecular effects of lithium can be the most important mechanisms of lithium that also alter neural transmissions including dopaminergic signaling in mesolimbic and mesocortical pathways.
Collapse
|
4
|
Juza R, Musilek K, Mezeiova E, Soukup O, Korabecny J. Recent advances in dopamine D 2 receptor ligands in the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:55-211. [PMID: 36111795 DOI: 10.1002/med.21923] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Dopamine is a biologically active amine synthesized in the central and peripheral nervous system. This biogenic monoamine acts by activating five types of dopamine receptors (D1-5 Rs), which belong to the G protein-coupled receptor family. Antagonists and partial agonists of D2 Rs are used to treat schizophrenia, Parkinson's disease, depression, and anxiety. The typical pharmacophore with high D2 R affinity comprises four main areas, namely aromatic moiety, cyclic amine, central linker and aromatic/heteroaromatic lipophilic fragment. From the literature reviewed herein, we can conclude that 4-(2,3-dichlorophenyl), 4-(2-methoxyphenyl)-, 4-(benzo[b]thiophen-4-yl)-1-substituted piperazine, and 4-(6-fluorobenzo[d]isoxazol-3-yl)piperidine moieties are critical for high D2 R affinity. Four to six atoms chains are optimal for D2 R affinity with 4-butoxyl as the most pronounced one. The bicyclic aromatic/heteroaromatic systems are most frequently occurring as lipophilic appendages to retain high D2 R affinity. In this review, we provide a thorough overview of the therapeutic potential of D2 R modulators in the treatment of the aforementioned disorders. In addition, this review summarizes current knowledge about these diseases, with a focus on the dopaminergic pathway underlying these pathologies. Major attention is paid to the structure, function, and pharmacology of novel D2 R ligands, which have been developed in the last decade (2010-2021), and belong to the 1,4-disubstituted aromatic cyclic amine group. Due to the abundance of data, allosteric D2 R ligands and D2 R modulators from patents are not discussed in this review.
Collapse
Affiliation(s)
- Radomir Juza
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Mezeiova
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
5
|
Gantz E, Daniel Sharer J, McGrath TM. Diagnosis of Aromatic L-Amino Acid Decarboxylase (AADC) Deficiency via Epilepsy Gene Panel Screening in a Patient with Atypical Presentation. Child Neurol Open 2023. [DOI: 10.1177/2329048x231161027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
We describe an atypical presentation of a girl with aromatic L-amino acid decarboxylase (AADC) deficiency identified via a genetic testing program for children with epilepsy. At 21 months of age, she presented with poor head control, diffuse hypotonia, poor fixation, developmental delay, and dysphagia. She was lost to follow-up, then presented back at 3 years of age with staring spells and brief episodes of upward eye deviation. The diagnosis of unprovoked epilepsy allowed her to be included in a genetic testing program, which identified two heterozygous variants in the dopa decarboxylase (DCC) gene. Based on the genetic testing, plasma AADC enzyme activity and plasma 3-O-methyldopa results, a diagnosis of AADC deficiency was made when she was 4 years and 2 months of age. This case report shows that AADC deficiency can be the underlying diagnosis in patients with suspected epilepsy.
Collapse
Affiliation(s)
- Emily Gantz
- Division of Pediatric Neurology, Department of Pediatrics, University of Alabama, Birmingham, AL, USA
| | - J. Daniel Sharer
- Biochemical Genetics Laboratory, Department of Genetics, University of Alabama, Birmingham, AL, USA
| | - Tony M. McGrath
- Division of Pediatric Neurology, Department of Pediatrics, University of Alabama, Birmingham, AL, USA
| |
Collapse
|
6
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
7
|
Guler M, Kavak E, Kivrak A. Electrochemical Dopamine Sensor Based on Gold Nanoparticles Electrodeposited on a Polymer/Reduced Graphene Oxide-Modified Glassy Carbon Electrode. ANAL LETT 2021. [DOI: 10.1080/00032719.2021.1990310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Muhammet Guler
- Faculty of Science, Department of Chemistry, Van Yüzüncü Yıl University, Van, Turkey
| | - Emrah Kavak
- Faculty of Science, Department of Chemistry, Van Yüzüncü Yıl University, Van, Turkey
| | - Arif Kivrak
- Faculty of Sciences and Arts, Department of Chemistry, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
8
|
Du Z, Tian X, Ma M, Somlo S, Weinstein AM, Wang T. Restoration of proximal tubule flow-activated transport prevents cyst growth in polycystic kidney disease. JCI Insight 2021; 6:146041. [PMID: 33886508 PMCID: PMC8262298 DOI: 10.1172/jci.insight.146041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/21/2021] [Indexed: 01/11/2023] Open
Abstract
Flow-activated Na+ and HCO3– transport in kidney proximal tubules (PT) underlies relatively constant fractional reabsorption during changes in glomerular filtration rate (GFR) or glomerulotubular balance (GTB). In view of hypothesized connections of epithelial cilia to flow sensing, we examined flow-activated transport in 3 polycystic kidney disease–related mouse models based on inducible conditional KO of Pkd1, Pkd2, and Kif3a. PTs were harvested from mice after gene inactivation but prior to cyst formation, and flow-mediated PT transport was measured. We confirm that higher flow increased both Na+ and HCO3– absorption in control mice, and we observed that this flow effect was preserved in PTs of Pkd1–/– and Kif3a–/–mice. However, flow activation was absent in Pkd2+/– and Pkd2–/– PT. In heterozygous (Pkd2+/–) mice, a dopamine receptor 1 (DA1) antagonist (SCH23390) restored transport flow sensitivity. When given chronically, this same antagonist reduced renal cyst formation in Pkd2–/–, as evidenced by reduced kidney weight, BUN, and the cystic index, when compared with untreated mice. In contrast, SCH23390 did not prevent cyst formation in Pkd1–/– mice. These results indicate that Pkd2 is necessary for normal GTB and that restoration of flow-activated transport by DA1 antagonist can slow renal cyst formation in Pkd2–/– mice.
Collapse
Affiliation(s)
| | - Xin Tian
- Department of Internal Medicine (Nephrology), Yale University, New Haven, Connecticut, USA
| | - Ming Ma
- Department of Internal Medicine (Nephrology), Yale University, New Haven, Connecticut, USA
| | - Stefan Somlo
- Department of Internal Medicine (Nephrology), Yale University, New Haven, Connecticut, USA
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, New York, USA
| | | |
Collapse
|
9
|
Sussman CR, Wang X, Chebib FT, Torres VE. Modulation of polycystic kidney disease by G-protein coupled receptors and cyclic AMP signaling. Cell Signal 2020; 72:109649. [PMID: 32335259 DOI: 10.1016/j.cellsig.2020.109649] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/11/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a systemic disorder associated with polycystic liver disease (PLD) and other extrarenal manifestations, the most common monogenic cause of end-stage kidney disease, and a major burden for public health. Many studies have shown that alterations in G-protein and cAMP signaling play a central role in its pathogenesis. As for many other diseases (35% of all approved drugs target G-protein coupled receptors (GPCRs) or proteins functioning upstream or downstream from GPCRs), treatments targeting GPCR have shown effectiveness in slowing the rate of progression of ADPKD. Tolvaptan, a vasopressin V2 receptor antagonist is the first drug approved by regulatory agencies to treat rapidly progressive ADPKD. Long-acting somatostatin analogs have also been effective in slowing the rates of growth of polycystic kidneys and liver. Although no treatment has so far been able to prevent the development or stop the progression of the disease, these encouraging advances point to G-protein and cAMP signaling as a promising avenue of investigation that may lead to more effective and safe treatments. This will require a better understanding of the relevant GPCRs, G-proteins, cAMP effectors, and of the enzymes and A-kinase anchoring proteins controlling the compartmentalization of cAMP signaling. The purpose of this review is to provide an overview of general GPCR signaling; the function of polycystin-1 (PC1) as a putative atypical adhesion GPCR (aGPCR); the roles of PC1, polycystin-2 (PC2) and the PC1-PC2 complex in the regulation of calcium and cAMP signaling; the cross-talk of calcium and cAMP signaling in PKD; and GPCRs, adenylyl cyclases, cyclic nucleotide phosphodiesterases, and protein kinase A as therapeutic targets in ADPKD.
Collapse
Affiliation(s)
- Caroline R Sussman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
10
|
Banday AA, Diaz AD, Lokhandwala M. Kidney dopamine D 1-like receptors and angiotensin 1-7 interaction inhibits renal Na + transporters. Am J Physiol Renal Physiol 2019; 317:F949-F956. [PMID: 31411069 DOI: 10.1152/ajprenal.00135.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The role of dopamine D1-like receptors (DR) in the regulation of renal Na+ transporters, natriuresis, and blood pressure is well established. However, the involvement of the angiotensin 1-7 (ANG 1-7)-Mas receptor in the regulation of Na+ balance and blood pressure is not clear. The present study aimed to investigate the hypothesis that ANG 1-7 can regulate Na+ homeostasis by modulating the renal dopamine system. Sprague-Dawley rats were infused with saline alone (vehicle) or saline with ANG 1-7, ANG 1-7 antagonist A-779, DR agonist SKF38393, and antagonist SCH23390. Infusion of ANG 1-7 caused significant natriuresis and diuresis compared with saline alone. Both natriuresis and diuresis were blocked by A-779 and SCH23390. SKF38393 caused a significant, SCH23390-sensitive natriuresis and diuresis, and A-779 had no effect on the SKF38393 response. Concomitant infusion of ANG 1-7 and SKF38393 did not show a cumulative effect compared with either agonist alone. Treatment of renal proximal tubules with ANG 1-7 or SKF38393 caused a significant decrease in Na+-K+-ATPase and Na+/H+ exchanger isoform 3 activity. While SCH23390 blocked both ANG 1-7- and SKF38393-induced inhibition, the DR response was not sensitive to A-779. Additionally, ANG 1-7 activated PKG, enhanced tyrosine hydroxylase activity via Ser40 phosphorylation, and increased renal dopamine production. These data suggest that ANG 1-7, via PKG, enhances tyrosine hydroxylase activity, which increases renal dopamine production and activation of DR and subsequent natriuresis. This study provides evidence for a unidirectional functional interaction between two G protein-coupled receptors to regulate renal Na+ transporters and induce natriuresis.
Collapse
Affiliation(s)
- Anees A Banday
- University of Houston, College of Pharmacy, Heart and Kidney Institute, Houston, Texas
| | - Andrea Diaz Diaz
- University of Houston, College of Pharmacy, Heart and Kidney Institute, Houston, Texas.,School of Pharmacy, University College Cork, Cork, Ireland
| | - Mustafa Lokhandwala
- University of Houston, College of Pharmacy, Heart and Kidney Institute, Houston, Texas
| |
Collapse
|
11
|
Taveira-da-Silva R, da Silva Sampaio L, Vieyra A, Einicker-Lamas M. L-Tyr-Induced Phosphorylation of Tyrosine Hydroxylase at Ser40: An Alternative Route for Dopamine Synthesis and Modulation of Na+/K+-ATPase in Kidney Cells. Kidney Blood Press Res 2019; 44:1-11. [PMID: 30808844 DOI: 10.1159/000497806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Dopamine (DA) is a natriuretic hormone that inhibits renal sodium reabsorption, being Angiotensin II (Ang II) its powerful counterpart. These two systems work together to maintain sodium homeostasis and consequently, the blood pressure (BP) within normal limits. We hypothesized that L-tyrosine (L-tyr) or L-dihydroxyphenylalanine (L-dopa) could inhibit the Na+/K+-ATPase activity. We also evaluated whether L-tyr treatment modulates Tyrosine Hydroxylase (TH). METHODS Experiments involved cultured LLCPK1 cells treated with L-tyr or L-dopa for 30 minutes a 37°C. In experiments on the effect of Dopa Descarboxylase (DDC) inhibition, cells were pre incubated for 15 minutes with 3-Hydroxybenzylhydrazine dihydrochloride (HBH), and them L-dopa was added for 30 minutes. Na+/K+-ATPase activity was quantified colorimetrically. We used immunoblotting and immunocytochemistry to identify the enzymes TH, DDC and the dopamine receptor D1R in LLCPK1 cells. TH activity was accessed by immunoblotting (increase in the phosphorylation). TH and DDC activities were also evaluated by the modulation of the Na+/K+-ATPase activity, which can be ascribed to the synthesis of dopamine. RESULTS LLCPK1 cells express the required machinery for DA synthesis: the enzymes TH, and (DDC) as well as its receptor D1R, were detected in control steady state cells. Cells treated with L-tyr or L-dopa showed an inhibition of the basolateral Na+/K+-ATPase activity. We can assume that DA formed in the cytoplasm from L-tyr or L-dopa led to inhibition of the Na+/K+-ATPase activity compared to control. L-tyr treatment increases TH phosphorylation at Ser40 by 100%. HBH, a specific DDC inhibitor; BCH, a LAT2 inhibitor; and Sch 23397, a specific D1R antagonist, totally suppressed the inhibition of Na+/K+-ATPase activity due to L-dopa or L-tyr administration, as indicated in the figures. CONCLUSION The results indicate that DA formed mainly from luminal L-tyr or L-dopa uptake by LAT2, can inhibit the Na+/K+-ATPase. In addition, our results showed for the very first time that TH activity is also significantly increased when the cells were exposed to L-tyr.
Collapse
Affiliation(s)
| | - Luzia da Silva Sampaio
- Carlos Chagas Filho Biophysics Institute, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Biophysics Institute, Rio de Janeiro Federal University, Rio de Janeiro, Brazil.,National Center for Structural Biology and Bio Imaging (CENABIO), Rio de Janeiro Federal University, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine (REGENERA), Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Carlos Chagas Filho Biophysics Institute, Rio de Janeiro Federal University, Rio de Janeiro, Brazil,
| |
Collapse
|
12
|
Klein MO, Battagello DS, Cardoso AR, Hauser DN, Bittencourt JC, Correa RG. Dopamine: Functions, Signaling, and Association with Neurological Diseases. Cell Mol Neurobiol 2019; 39:31-59. [PMID: 30446950 DOI: 10.1007/s10571-018-0632-3] [Citation(s) in RCA: 497] [Impact Index Per Article: 99.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
Abstract
The dopaminergic system plays important roles in neuromodulation, such as motor control, motivation, reward, cognitive function, maternal, and reproductive behaviors. Dopamine is a neurotransmitter, synthesized in both central nervous system and the periphery, that exerts its actions upon binding to G protein-coupled receptors. Dopamine receptors are widely expressed in the body and function in both the peripheral and the central nervous systems. Dopaminergic signaling pathways are crucial to the maintenance of physiological processes and an unbalanced activity may lead to dysfunctions that are related to neurodegenerative diseases. Unveiling the neurobiology and the molecular mechanisms that underlie these illnesses may contribute to the development of new therapies that could promote a better quality of life for patients worldwide. In this review, we summarize the aspects of dopamine as a catecholaminergic neurotransmitter and discuss dopamine signaling pathways elicited through dopamine receptor activation in normal brain function. Furthermore, we describe the potential involvement of these signaling pathways in evoking the onset and progression of some diseases in the nervous system, such as Parkinson's, Schizophrenia, Huntington's, Attention Deficit and Hyperactivity Disorder, and Addiction. A brief description of new dopaminergic drugs recently approved and under development treatments for these ailments is also provided.
Collapse
Affiliation(s)
- Marianne O Klein
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Daniella S Battagello
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - Ariel R Cardoso
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil
| | - David N Hauser
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Jackson C Bittencourt
- Laboratory of Chemical Neuroanatomy, Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, 05508-000, Brazil.
- Center for Neuroscience and Behavior, Institute of Psychology, USP, São Paulo, Brazil.
| | - Ricardo G Correa
- Center for Translational Neuroscience, Sanford Burnham Prebys (SBP) Medical Discovery Institute, 10901 North Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
13
|
Hodorová I, Rybárová S, Solár P, Benický M, Rybár D, Kováčová Z, Mihalik J. Monoamine Oxidase B in Renal Cell Carcinoma. Med Sci Monit 2018; 24:5422-5426. [PMID: 30076780 PMCID: PMC6088511 DOI: 10.12659/msm.909507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Studies on monoamine oxidase B (MAO-B) expression in renal cell carcinoma (RCC) are lacking. This study focused on the immunohistochemical evaluation of MAO-B in RCC. MATERIAL AND METHODS Sixty-three RCC samples were compared on basic clinical and histopathological parameters, including histopathological type and tumor grade. RCC samples were divided according to the histopathological type into 2 groups: conventional type (51 samples) and other types (12 samples). For MAO-B detection, a standard immunohistochemical procedure was employed. RESULTS In healthy kidney samples, MAO-B was detected predominantly in tubules. Fifty-two cancer tissue samples were MAO-B negative and 11 tissue samples were MAO-B low positive. Enzymes were detected only in the cytoplasm. We did not find any significant correlation between the percentage of positive MAO-B specimens and nuclear grade. Additionally, Fisher's test did not reveal any difference in numbers of positive and negative MAO-B samples between the 2 RCC types (P>0.05). CONCLUSIONS From our results, it was clear that MAO-B expression played no significant role in stimulation of renal cancer development. We found that MAO-B occurred only in 19% of kidney tumors and that the positivity of protein expression was low. Moreover, it seems that the disappearance of this enzyme in RCC is a consequence of replacement of healthy tissue by cancer cells. On the other hand, one can assume that the loss of MAO-B expression could be associated with severe pathological processes in the kidney.
Collapse
Affiliation(s)
- Ingrid Hodorová
- Department of Anatomy, P.J. Šafárik University, Faculty of Medicine, Košice, Slovakia
| | - Silvia Rybárová
- Department of Anatomy, P.J. Šafárik University, Faculty of Medicine, Košice, Slovakia
| | - Peter Solár
- Department of Medical Biology, P.J. Šafárik University, Faculty of Medicine, Košice, Slovakia
| | - Marián Benický
- Department of Pathology, P.J. Šafárik University, Faculty of Medicine, Košice, Slovakia
| | - Dušan Rybár
- Department of Anesthesiology and Intensive Medicine, P.J. Šafárik University, Faculty of Medicine and VUSCH a.s., Košice, Slovakia
| | - Zuzana Kováčová
- Department of Anatomy, P.J. Šafárik University, Faculty of Medicine, Košice, Slovakia
| | - Jozef Mihalik
- Department of Anatomy, P.J. Šafárik University, Faculty of Medicine, Košice, Slovakia
| |
Collapse
|
14
|
Rukavina Mikusic NL, Kouyoumdzian NM, Uceda A, Del Mauro JS, Pandolfo M, Gironacci MM, Puyó AM, Toblli JE, Fernández BE, Choi MR. Losartan prevents the imbalance between renal dopaminergic and renin angiotensin systems induced by fructose overload. l-Dopa/dopamine index as new potential biomarker of renal dysfunction. Metabolism 2018; 85:271-285. [PMID: 29727629 DOI: 10.1016/j.metabol.2018.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/28/2018] [Accepted: 04/25/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND The renin angiotensin system (RAS) and the renal dopaminergic system (RDS) act as autocrine and paracrine systems to regulate renal sodium management and inflammation and their alterations have been associated to hypertension and renal damage. Nearly 30-50% of hypertensive patients have insulin resistance (IR), with a strong correlation between hyperinsulinemia and microalbuminuria. OBJECTIVE The aim of this study was to demonstrate the existence of an imbalance between RAS and RDS associated to IR, hypertension and kidney damage induced by fructose overload (FO), as well as to establish their prevention, by pharmacological inhibition of RAS with losartan. MATERIALS/METHODS Ninety-six male Sprague-Dawley rats were randomly divided into four groups and studied at 4, 8 and 12 weeks: control group (C4, C8 and C12; tap water to drink); fructose-overloaded group (F4, F8 and F12; 10% w/v fructose solution to drink); losartan-treated control (L) group (L4, L8 and L12; losartan 30 mg/kg/day, in drinking water); and fructose-overloaded plus losartan group (F + L4, F + L8 and F + L12, in fructose solution). RESULTS FO induced metabolic and hemodynamic alterations as well as an imbalance between RAS and RDS, characterized by increased renal angiotensin II levels and AT1R overexpression, reduced urinary excretion of dopamine, increased excretion of l-dopa (increased l-dopa/dopamine index) and down-regulation of D1R and tubular dopamine transporters OCT-2, OCT-N1 and total OCTNs. This imbalance was accompanied by an overexpression of renal tubular Na+, K+-ATPase, pro-inflammatory (NF-kB, TNF-α, IL-6) and pro-fibrotic (TGF-β1 and collagen) markers and by renal damage (microalbuminuria and reduced nephrin expression). Losartan prevented the metabolic and hemodynamic alterations induced by FO from week 4. Increased urinary l-dopa/dopamine index and decreased D1R renal expression associated to FO were also prevented by losartan since week 4. The same pattern was observed for renal expression of OCTs/OCTNs, Na+, K+-ATPase, pro-inflammatory and pro-fibrotic markers from week 8. The appearance of microalbuminuria and reduced nephrin expression was prevented by losartan at week 12. CONCLUSION The results of this study provide new insight regarding the mechanisms by which a pro-hypertensive and pro-inflammatory system, such as RAS, downregulates another anti-hypertensive and anti-inflammatory system such as RDS. Additionally, we propose the use of l-dopa/dopamine index as a biochemical marker of renal dysfunction in conditions characterized by sodium retention, IR and/or hypertension, and as a predictor of response to treatment and follow-up of these processes.
Collapse
Affiliation(s)
- Natalia Lucía Rukavina Mikusic
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina.
| | - Nicolás Martín Kouyoumdzian
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Ana Uceda
- Hospital Alemán, Laboratorio de Medicina Experimental, Av Pueyrredón 1640, C1118AAT CABA, Buenos Aires, Argentina
| | - Julieta Sofía Del Mauro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Farmacología, Cátedra de Farmacología, Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Marcela Pandolfo
- Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Mariela Mercedes Gironacci
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica, Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Ana María Puyó
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Junín 956, C1113AAD CABA, Buenos Aires, Argentina
| | - Jorge Eduardo Toblli
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Hospital Alemán, Laboratorio de Medicina Experimental, Av Pueyrredón 1640, C1118AAT CABA, Buenos Aires, Argentina
| | - Belisario Enrique Fernández
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina; Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Av. Gral Las Heras 2191, C1127AAD CABA, Buenos Aires, Argentina
| | - Marcelo Roberto Choi
- CONICET, Universidad de Buenos Aires, Instituto de Investigaciones Cardiológicas (ININCA), Marcelo T. de Alvear 2270, C1122AAJ City of Buenos Aires (CABA), Buenos Aires, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Junín 956, C1113AAD CABA, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Junín 956, C1113AAD CABA, Buenos Aires, Argentina; Instituto Universitario de Ciencias de la Salud, Fundación H.A. Barceló, Av. Gral Las Heras 2191, C1127AAD CABA, Buenos Aires, Argentina
| |
Collapse
|
15
|
Taub M. Gene Level Regulation of Na,K-ATPase in the Renal Proximal Tubule Is Controlled by Two Independent but Interacting Regulatory Mechanisms Involving Salt Inducible Kinase 1 and CREB-Regulated Transcriptional Coactivators. Int J Mol Sci 2018; 19:E2086. [PMID: 30021947 PMCID: PMC6073390 DOI: 10.3390/ijms19072086] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022] Open
Abstract
For many years, studies concerning the regulation of Na,K-ATPase were restricted to acute regulatory mechanisms, which affected the phosphorylation of Na,K-ATPase, and thus its retention on the plasma membrane. However, in recent years, this focus has changed. Na,K-ATPase has been established as a signal transducer, which becomes part of a signaling complex as a consequence of ouabain binding. Na,K-ATPase within this signaling complex is localized in caveolae, where Na,K-ATPase has also been observed to regulate Inositol 1,4,5-Trisphosphate Receptor (IP3R)-mediated calcium release. This latter association has been implicated as playing a role in signaling by G Protein Coupled Receptors (GPCRs). Here, the consequences of signaling by renal effectors that act via such GPCRs are reviewed, including their regulatory effects on Na,K-ATPase gene expression in the renal proximal tubule (RPT). Two major types of gene regulation entail signaling by Salt Inducible Kinase 1 (SIK1). On one hand, SIK1 acts so as to block signaling via cAMP Response Element (CRE) Binding Protein (CREB) Regulated Transcriptional Coactivators (CRTCs) and on the other hand, SIK1 acts so as to stimulate signaling via the Myocyte Enhancer Factor 2 (MEF2)/nuclear factor of activated T cell (NFAT) regulated genes. Ultimate consequences of these pathways include regulatory effects which alter the rate of transcription of the Na,K-ATPase β1 subunit gene atp1b1 by CREB, as well as by MEF2/NFAT.
Collapse
Affiliation(s)
- Mary Taub
- Biochemistry Dept., Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 955 Main Street, Suite 4902, Buffalo, NY 14203, USA.
| |
Collapse
|
16
|
Dumont E, De Bleye C, Cailletaud J, Sacré PY, Van Lerberghe PB, Rogister B, Rance GA, Aylott JW, Hubert P, Ziemons E. Development of a SERS strategy to overcome the nanoparticle stabilisation effect in serum-containing samples: Application to the quantification of dopamine in the culture medium of PC-12 cells. Talanta 2018; 186:8-16. [PMID: 29784422 DOI: 10.1016/j.talanta.2018.04.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/02/2018] [Accepted: 04/08/2018] [Indexed: 12/19/2022]
Abstract
The analysis of serum samples by surface-enhanced Raman spectroscopy (SERS) has gained ground over the last few years. However, the stabilisation of colloids by the proteins contained in these samples has restricted their use in common practice, unless antibodies or aptamers are used. Therefore, this work was dedicated to the development of a SERS methodology allowing the analysis of serum samples in a simple and easy-to-implement way. This approach was based on the pre-aggregation of the colloid with a salt solution. Gold nanoparticles (AuNPs) were used as the SERS substrate and, owing to its physiopathological importance, dopamine was chosen as a model to implement the SERS approach. The presence of this neurotransmitter could be determined in the concentration range 0.5-50 ppm (2.64-264 µM) in the culture medium of PC-12 cells, with a R2 of 0.9874, and at even lower concentrations (0.25 ppm, 1.32 µM) in another matrix containing fewer proteins. Moreover, the effect of calcium and potassium on the dopamine exocytosis from PC-12 cells was studied. Calcium was shown to have a predominant and dose-dependant effect. Finally, PC-12 cells were exposed to dexamethasone in order to increase their biosynthesis and release of dopamine. This increase was monitored with the developed SERS approach.
Collapse
Affiliation(s)
- E Dumont
- University of Liege (ULiege), CIRM, VibraSanté Hub, Department of Pharmacy, Laboratory of Pharmaceutical Analytical Chemistry, CHU, B36, B-4000 Liege, Belgium.
| | - C De Bleye
- University of Liege (ULiege), CIRM, VibraSanté Hub, Department of Pharmacy, Laboratory of Pharmaceutical Analytical Chemistry, CHU, B36, B-4000 Liege, Belgium
| | - J Cailletaud
- University of Liege (ULiege), CIRM, VibraSanté Hub, Department of Pharmacy, Laboratory of Pharmaceutical Analytical Chemistry, CHU, B36, B-4000 Liege, Belgium
| | - P-Y Sacré
- University of Liege (ULiege), CIRM, VibraSanté Hub, Department of Pharmacy, Laboratory of Pharmaceutical Analytical Chemistry, CHU, B36, B-4000 Liege, Belgium
| | - P-B Van Lerberghe
- University of Liege (ULiege), GIGA-Neurosciences, Laboratory of Developmental Neurobiology, CHU, B36, B-4000 Liege, Belgium
| | - B Rogister
- University of Liege (ULiege), GIGA-Neurosciences, Laboratory of Nervous System Disorders and Therapy, CHU, B36, B-4000 Liege, Belgium; Department of Neurology, CHU of Liège, B-4000 Liege, Belgium
| | - G A Rance
- University of Nottingham, Nanoscale and Microscale Research Centre, University Park, NG7 2RD Nottingham, United Kingdom
| | - J W Aylott
- University of Nottingham, School of Pharmacy, Laboratory of Biophysics and Surface Analysis, University Park, NG7 2RD Nottingham, United Kingdom
| | - Ph Hubert
- University of Liege (ULiege), CIRM, VibraSanté Hub, Department of Pharmacy, Laboratory of Pharmaceutical Analytical Chemistry, CHU, B36, B-4000 Liege, Belgium
| | - E Ziemons
- University of Liege (ULiege), CIRM, VibraSanté Hub, Department of Pharmacy, Laboratory of Pharmaceutical Analytical Chemistry, CHU, B36, B-4000 Liege, Belgium
| |
Collapse
|
17
|
Wang T, Weinbaum S, Weinstein AM. Regulation of glomerulotubular balance: flow-activated proximal tubule function. Pflugers Arch 2017; 469:643-654. [PMID: 28271233 DOI: 10.1007/s00424-017-1960-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 02/15/2017] [Accepted: 02/17/2017] [Indexed: 12/23/2022]
Abstract
The purpose of this review is to summarize our knowledge and understanding of the physiological importance and the mechanisms underlying flow-activated proximal tubule transport. Since the earliest micropuncture studies of mammalian proximal tubule, it has been recognized that tubular flow is an important regulator of sodium, potassium, and acid-base transport in the kidney. Increased fluid flow stimulates Na+ and HCO3- absorption in the proximal tubule via stimulation of Na/H-exchanger isoform 3 (NHE3) and H+-ATPase. In the proximal tubule, brush border microvilli are the major flow sensors, which experience changes in hydrodynamic drag and bending moment as luminal flow velocity changes and which transmit the force of altered flow to cytoskeletal structures within the cell. The signal to NHE3 depends upon the integrity of the actin cytoskeleton; the signal to the H+-ATPase depends upon microtubules. We have demonstrated that alterations in fluid drag impact tubule function by modulating ion transporter availability within the brush border membrane of the proximal tubule. Beyond that, there is evidence that transporter activity within the peritubular membrane is also modulated by luminal flow. Secondary messengers that regulate the flow-mediated tubule function have also been delineated. Dopamine blunts the responsiveness of proximal tubule transporters to changes in luminal flow velocity, while a DA1 antagonist increases flow sensitivity of solute reabsorption. IP3 receptor-mediated intracellular Ca2+ signaling is critical to transduction of microvillus drag. In this review, we summarize our findings of the regulatory mechanism of flow-mediated Na+ and HCO3- transport in the proximal tubule and review available information about flow sensing and regulatory mechanism of glomerulotubular balance.
Collapse
Affiliation(s)
- Tong Wang
- Department of Cellular and Molecular Physiology, Yale University, 333 Cedar Street, New Haven, CT, 06520, USA.
| | - Sheldon Weinbaum
- Department of Biomedical Engineering, City College of New York, CUNY, New York, NY, USA
| | - Alan M Weinstein
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
18
|
Ibarra ME, Albertoni Borghese MF, Majowicz MP, Ortiz MC, Loidl F, Rey-Funes M, Di Ciano LA, Ibarra FR. Concerted regulation of renal plasma flow and glomerular filtration rate by renal dopamine and NOS I in rats on high salt intake. Physiol Rep 2017; 5:e13202. [PMID: 28351967 PMCID: PMC5371567 DOI: 10.14814/phy2.13202] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/05/2017] [Accepted: 02/20/2017] [Indexed: 11/24/2022] Open
Abstract
Under high sodium intake renal dopamine (DA) increases while NOS I expression in macula densa cells (MD) decreases. To explore whether renal DA and NOS I, linked to natriuresis and to the stability of the tubuloglomerular feedback, respectively, act in concert to regulate renal plasma flow (RPF) and glomerular filtration rate (GFR). Male Wistar rats were studied under a normal sodium intake (NS, NaCl 0.24%) or a high sodium intake (HS, NaCl 1% in drinking water) during the 5 days of the study. For the last two days, the specific D1-like receptor antagonist SCH 23390 (1 mg kg bwt-1 day-1, sc) or a vehicle was administered. HS intake increased natriuresis, diuresis, and urinary DA while it decreased cortical NOS I expression (P < 0.05 vs. NS), Nicotinamide adenine dinucleotide phosphate diaphorase (NADPH-d) activity in MD (P < 0.001 vs. NS) and cortical nitrates+nitrites (NOx) production (NS 2.04 ± 0.22 vs. HS 1.28 ± 0.10 nmol mg protein-1, P < 0.01). Treatment with SCH 23390 to rats on HS sharply decreased hydroelectrolyte excretion (P < 0.001 vs. HS) while NOS I expression, NADPH-d activity and NOx production increased (P < 0.05 vs. HS for NOS I and P < 0.001 vs. HS for NADPH-d and NOx). SCH 23390 increased RPF and GFR in HS rats (P < 0.01 HS+SCH vs. HS). It did not cause variations in NS rats. Results indicate that when NS intake is shifted to a prolonged high sodium intake, renal DA through the D1R, and NOS I in MD cells act in concert to regulate RPF and GFR to stabilize the delivery of NaCl to the distal nephron.
Collapse
Affiliation(s)
- Mariano E Ibarra
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria F Albertoni Borghese
- Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mónica P Majowicz
- Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María C Ortiz
- Cátedra de Biología Celular y Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fabián Loidl
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Manuel Rey-Funes
- Laboratorio de Neuropatología Experimental, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luis A Di Ciano
- Laboratorio de Riñón, Instituto de Investigaciones Médicas A. Lanari Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Fernando R Ibarra
- Laboratorio de Riñón, Instituto de Investigaciones Médicas A. Lanari Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Ciencias Fisiológicas, Facultad de Medicina Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
19
|
Tang L, Zheng S, Ren H, He D, Zeng C, Wang WE. Activation of angiotensin II type 1 receptors increases D 4 dopamine receptor expression in rat renal proximal tubule cells. Hypertens Res 2017; 40:652-657. [PMID: 28230199 DOI: 10.1038/hr.2017.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 12/22/2022]
Abstract
Both the dopaminergic and renin-angiotensin systems play important roles in the regulation of blood pressure. Our previous study showed that the stimulation of dopaminergic D4 receptors reduced angiotensin II type 1 (AT1) receptor expression in renal proximal tubule (RPT) cells. In this study, we tested whether AT1 receptors, in return, would regulate D4 receptor expression and function in RPT cells. Expression of the D4 receptor from Wistar-Kyoto (WKY) or spontaneously hypertensive rats (SHRs) RPT cells and renal cortex tissues were determined by western blot, and Na+-K+ ATPase activity was determined using an enzyme assay. Urine volume and urine sodium of WKY rats and SHRs treated with or without D4 receptor stimulation were measured. Thus, activation of AT1 receptors with angiotensin II (Ang II) increased D4 receptor protein expression in RPT cells, and this increase was blocked by nicardipine, a calcium influx blocker. The D4 receptor agonist PD168077 inhibited Na+-K+ ATPase activity in WKY RPT cells but not in SHR RPT cells. Ang II pre-treatment promoted D4 receptor-mediated inhibition of Na+-K+ ATPase in RPT cells in WKY rats but not in SHRs. Meanwhile, Ang II pre-treatment augmented the natriuretic effect of PD168077 in WKY rats but not in SHRs. In conclusion, AT1 stimulation can regulate the expression and natriuretic function of dopaminergic D4 receptors in RPT cells and might be involved in the pathogenesis of essential hypertension.
Collapse
Affiliation(s)
- Luxun Tang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Duofen He
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| | - Wei Eric Wang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Institute of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, PR China.,Chongqing Cardiovascular Clinical Research Center, Daping Hospital, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
20
|
Schulze U, Brast S, Grabner A, Albiker C, Snieder B, Holle S, Schlatter E, Schröter R, Pavenstädt H, Herrmann E, Lambert C, Spoden GA, Florin L, Saftig P, Ciarimboli G. Tetraspanin CD63 controls basolateral sorting of organic cation transporter 2 in renal proximal tubules. FASEB J 2016; 31:1421-1433. [DOI: 10.1096/fj.201600901r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/12/2016] [Indexed: 12/19/2022]
Affiliation(s)
- Ulf Schulze
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Sabine Brast
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Alexander Grabner
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Christian Albiker
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Beatrice Snieder
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Svenja Holle
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Eberhard Schlatter
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Rita Schröter
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Hermann Pavenstädt
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| | - Edwin Herrmann
- Klinik und Poliklinik für UrologieWestfälische Wilhelms–Universität Münster Germany
| | - Carsten Lambert
- Institut für Medizinische Mikrobiologie und HygieneJohannes Gutenberg–Universität Mainz Germany
| | - Gilles A. Spoden
- Institut für Medizinische Mikrobiologie und HygieneJohannes Gutenberg–Universität Mainz Germany
| | - Luise Florin
- Institut für Medizinische Mikrobiologie und HygieneJohannes Gutenberg–Universität Mainz Germany
| | - Paul Saftig
- Biochemisches Institut, Medizinische FakultätChristian‐Albrechts–Universität Kiel Germany
| | - Giuliano Ciarimboli
- Medizinische Klinik D, Experimentelle NephrologieWestfälische Wilhelms–Universität Münster Germany
| |
Collapse
|
21
|
Dopamine regulates renal osmoregulation during hyposaline stress via DRD1 in the spotted scat (Scatophagus argus). Sci Rep 2016; 6:37535. [PMID: 27857228 PMCID: PMC5114590 DOI: 10.1038/srep37535] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/01/2016] [Indexed: 01/11/2023] Open
Abstract
Dopamine is an important regulator of renal natriuresis and is critical for the adaptation of many animals to changing environmental salinity. However, the molecular mechanisms through which dopamine promotes this adaptation remain poorly understood. We studied the effects of dopamine on renal hypo-osmoregulation in the euryhaline fish Scatophagus argus (S. argus) during abrupt transfer from seawater (SW) to freshwater (FW). Following the transfer, serum dopamine concentration was decreased, and dopamine activated expression of the dopamine receptor 1 (designated SaDRD1) in the kidney, triggering the osmoregulatory signaling cascade. SaDRD1 protein is expressed in the renal proximal tubule cells in vivo, and is localized to the cell membrane of renal primary cells in vitro. Knockdown of SaDRD1 mRNA by siRNA significantly increased Na+/K+-ATPase (NKA) activity in cultured renal primary cells in vitro, suggesting that expression of SaDRD1 may oppose the activity of NKA. We demonstrate that exogenous dopamine enhances the response of NKA to hyposaline stress after transferring primary renal cells from isosmotic medium to hypoosmotic medium. Our results indicate that dopamine regulation via SaDRD1 ignited the renal dopaminergic system to balance the osmotic pressure through inhibiting NKA activity, providing a new perspective on the hyposaline adaptation of fish.
Collapse
|
22
|
Rukavina Mikusic NL, Kouyoumdzian NM, Rouvier E, Gironacci MM, Toblli JE, Fernández BE, Choi MR. Regulation of Dopamine Uptake by Vasoactive Peptides in the Kidney. SCIENTIFICA 2016; 2016:6302376. [PMID: 27635280 PMCID: PMC5011208 DOI: 10.1155/2016/6302376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 06/19/2016] [Accepted: 07/03/2016] [Indexed: 06/06/2023]
Abstract
Considering the key role of renal dopamine in tubular sodium handling, we hypothesized that c-type natriuretic peptide (CNP) and Ang-(1-7) may regulate renal dopamine availability in tubular cells, contributing to Na(+), K(+)-ATPase inhibition. Present results show that CNP did not affect either (3)H-dopamine uptake in renal tissue or Na(+), K(+)-ATPase activity; meanwhile, Ang-(1-7) was able to increase (3)H-dopamine uptake and decreased Na(+), K(+)-ATPase activity in renal cortex. Ang-(1-7) and dopamine together decreased further Na(+), K(+)-ATPase activity showing an additive effect on the sodium pump. In addition, hydrocortisone reversed Ang-(1-7)-dopamine overinhibition on the enzyme, suggesting that this inhibition is closely related to Ang-(1-7) stimulation on renal dopamine uptake. Both anantin and cANP (4-23-amide) did not modify CNP effects on (3)H-dopamine uptake by tubular cells. The Mas receptor antagonist, A-779, blocked the increase elicited by Ang-(1-7) on (3)H-dopamine uptake. The stimulatory uptake induced by Ang-(1-7) was even more pronounced in the presence of losartan, suggesting an inhibitory effect of Ang-(1-7) on AT1 receptors on (3)H-dopamine uptake. By increasing dopamine bioavailability in tubular cells, Ang-(1-7) enhances Na(+), K(+)-ATPase activity inhibition, contributing to its natriuretic and diuretic effects.
Collapse
Affiliation(s)
- N. L. Rukavina Mikusic
- Instituto de Investigaciones Cardiológicas ININCA, UBA-CONICET, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
| | - N. M. Kouyoumdzian
- Instituto de Investigaciones Cardiológicas ININCA, UBA-CONICET, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
| | - E. Rouvier
- Instituto de Investigaciones Cardiológicas ININCA, UBA-CONICET, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
- Cátedras de Anatomía e Histología, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
| | - M. M. Gironacci
- Cátedras de Química Biológica, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
| | - J. E. Toblli
- Instituto de Investigaciones Cardiológicas ININCA, UBA-CONICET, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
- Laboratorio de Medicina Experimental, Hospital Alemán, Buenos Aires, Argentina
| | - B. E. Fernández
- Instituto de Investigaciones Cardiológicas ININCA, UBA-CONICET, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
| | - M. R. Choi
- Instituto de Investigaciones Cardiológicas ININCA, UBA-CONICET, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
- Cátedras de Anatomía e Histología, Facultad de Farmacia y Bioquímica, UBA, Buenos Aires, Argentina
| |
Collapse
|
23
|
Kouyoumdzian NM, Rukavina Mikusic NL, Kravetz MC, Lee BM, Carranza A, Del Mauro JS, Pandolfo M, Gironacci MM, Gorzalczany S, Toblli JE, Fernández BE, Choi MR. Atrial Natriuretic Peptide Stimulates Dopamine Tubular Transport by Organic Cation Transporters: A Novel Mechanism to Enhance Renal Sodium Excretion. PLoS One 2016; 11:e0157487. [PMID: 27392042 PMCID: PMC4938554 DOI: 10.1371/journal.pone.0157487] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 05/30/2016] [Indexed: 01/11/2023] Open
Abstract
The aim of this study was to demonstrate the effects of atrial natriuretic peptide (ANP) on organic cation transporters (OCTs) expression and activity, and its consequences on dopamine urinary levels, Na+, K+-ATPase activity and renal function. Male Sprague Dawley rats were infused with isotonic saline solution during 120 minutes and randomized in nine different groups: control, pargyline plus tolcapone (P+T), ANP, dopamine (DA), D-22, DA+D-22, ANP+D-22, ANP+DA and ANP+DA+D-22. Renal functional parameters were determined and urinary dopamine concentration was quantified by HPLC. Expression of OCTs and D1-receptor in membrane preparations from renal cortex tissues were determined by western blot and Na+, K+-ATPase activity was determined using in vitro enzyme assay. 3H-DA renal uptake was determined in vitro. Compared to P+T group, ANP and dopamine infusion increased diuresis, urinary sodium and dopamine excretion significantly. These effects were more pronounced in ANP+DA group and reversed by OCTs blockade by D-22, demonstrating that OCTs are implied in ANP stimulated-DA uptake and transport in renal tissues. The activity of Na+, K+-ATPase exhibited a similar fashion when it was measured in the same experimental groups. Although OCTs and D1-receptor protein expression were not modified by ANP, OCTs-dependent-dopamine tubular uptake was increased by ANP through activation of NPR-A receptor and protein kinase G as signaling pathway. This effect was reflected by an increase in urinary dopamine excretion, natriuresis, diuresis and decreased Na+, K+-ATPase activity. OCTs represent a novel target that links the activity of ANP and dopamine together in a common mechanism to enhance their natriuretic and diuretic effects.
Collapse
Affiliation(s)
- Nicolás M. Kouyoumdzian
- Cardiological Research Institute, National Scientific and Technical Research Council, Buenos Aires, Argentina
- Pathophysiology and Clinical Biochemistry Institute, Buenos Aires, Argentina
| | - Natalia L. Rukavina Mikusic
- Cardiological Research Institute, National Scientific and Technical Research Council, Buenos Aires, Argentina
- Pathophysiology and Clinical Biochemistry Institute, Buenos Aires, Argentina
| | - María C. Kravetz
- Department of Pharmacology, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Brenda M. Lee
- Department of General Surgery, Johns Hopkins Hospital, Baltimore, Maryland, United States of America
| | - Andrea Carranza
- Department of Pharmacology, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Julieta S. Del Mauro
- Department of Pharmacology, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Marcela Pandolfo
- Pathophysiology and Clinical Biochemistry Institute, Buenos Aires, Argentina
| | - Mariela M. Gironacci
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Susana Gorzalczany
- Department of Pharmacology, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Jorge E. Toblli
- Cardiological Research Institute, National Scientific and Technical Research Council, Buenos Aires, Argentina
- Deutsch Hospital, Buenos Aires, Argentina
| | - Belisario E. Fernández
- Cardiological Research Institute, National Scientific and Technical Research Council, Buenos Aires, Argentina
- Pathophysiology and Clinical Biochemistry Institute, Buenos Aires, Argentina
| | - Marcelo R. Choi
- Cardiological Research Institute, National Scientific and Technical Research Council, Buenos Aires, Argentina
- Pathophysiology and Clinical Biochemistry Institute, Buenos Aires, Argentina
- Department of Anatomy and Histology, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
- Department of Pathophysiology, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
24
|
Arystarkhova E. Beneficial Renal and Pancreatic Phenotypes in a Mouse Deficient in FXYD2 Regulatory Subunit of Na,K-ATPase. Front Physiol 2016; 7:88. [PMID: 27014088 PMCID: PMC4779850 DOI: 10.3389/fphys.2016.00088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/22/2016] [Indexed: 11/25/2022] Open
Abstract
The fundamental role of Na,K-ATPase in eukaryotic cells calls for complex and efficient regulation of its activity. Besides alterations in gene expression and trafficking, kinetic properties of the pump are modulated by reversible association with single span membrane proteins, the FXYDs. Seven members of the family are expressed in a tissue-specific manner, affecting pump kinetics in all possible permutations. This mini-review focuses on functional properties of FXYD2 studied in transfected cells, and on noteworthy and unexpected phenotypes discovered in a Fxyd2−∕− mouse. FXYD2, the gamma subunit, reduces activity of Na,K-ATPase either by decreasing affinity for Na+, or reducing Vmax. FXYD2 mRNA splicing and editing provide another layer for regulation of Na,K-ATPase. In kidney of knockouts, there was elevated activity for Na,K-ATPase and for NCC and NKCC2 apical sodium transporters. That should lead to sodium retention and hypertension, however, the mice were in sodium balance and normotensive. Adult Fxyd2−∕− mice also exhibited a mild pancreatic phenotype with enhanced glucose tolerance, elevation of circulating insulin, but no insulin resistance. There was an increase in beta cell proliferation and beta cell mass that correlated with activation of the PI3K-Akt pathway. The Fxyd2−∕− mice are thus in a highly desirable state: the animals are resistant to Na+ retention, and showed improved glucose control, i.e., they display favorable metabolic adaptations to protect against development of salt-sensitive hypertension and diabetes. Investigation of the mechanisms of these adaptations in the mouse has the potential to unveil a novel therapeutic FXYD2-dependent strategy.
Collapse
Affiliation(s)
- Elena Arystarkhova
- Laboratory of Membrane Biology, Neurosurgery, Massachusetts General Hospital Boston, MA, USA
| |
Collapse
|
25
|
Taub M, Garimella S, Kim D, Rajkhowa T, Cutuli F. Renal proximal tubule Na,K-ATPase is controlled by CREB-regulated transcriptional coactivators as well as salt-inducible kinase 1. Cell Signal 2015; 27:2568-78. [PMID: 26432356 PMCID: PMC4696386 DOI: 10.1016/j.cellsig.2015.09.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/17/2015] [Accepted: 09/28/2015] [Indexed: 01/11/2023]
Abstract
Sodium reabsorption by the kidney is regulated by locally produced natriuretic and anti-natriuretic factors, including dopamine and norepinephrine, respectively. Previous studies indicated that signaling events initiated by these natriuretic and anti-natriuretic factors achieve their effects by altering the phosphorylation of Na,K-ATPase in the renal proximal tubule, and that protein kinase A (PKA) and calcium-mediated signaling pathways are involved. The same signaling pathways also control the transcription of the Na,K-ATPase β subunit gene atp1b1 in renal proximal tubule cells. In this report, evidence is presented that (1) both the recently discovered cAMP-regulated transcriptional coactivators (CRTCs) and salt-inducible kinase 1 (SIK1) contribute to the transcriptional regulation of atp1b1 in renal proximal tubule (RPT) cells and (2) renal effectors, including norepinephrine, dopamine, prostaglandins, and sodium, play a role. Exogenously expressed CRTCs stimulate atp1b1 transcription. Evidence for a role of endogenous CRTCs includes the loss of transcriptional regulation of atp1b1 by a dominant-negative CRTC, as well as by a CREB mutant, with an altered CRTC binding site. In a number of experimental systems, SIK phosphorylates CRTCs, which are then sequestered in the cytoplasm, preventing their nuclear effects. Consistent with such a role of SIK in primary RPT cells, atp1b1 transcription increased in the presence of a dominant-negative SIK1, and in addition, regulation by dopamine, norepinephrine, and monensin was disrupted by a dominant-negative SIK1. These latter observations can be explained if SIK1 is phosphorylated and inactivated in the presence of these renal effectors. Our results support the hypothesis that Na,K-ATPase in the renal proximal tubule is regulated at the transcriptional level via SIK1 and CRTCs by renal effectors, in addition to the previously reported control of the phosphorylation of Na,K-ATPase.
Collapse
Affiliation(s)
- Mary Taub
- Biochemistry Department,School of Medicine and Biomedical SciencesUniversity at Buffalo,140 Farber Hall, 3435 Main Street,Buffalo, NY 14214, USA.
| | - Sudha Garimella
- Biochemistry Department,School of Medicine and Biomedical SciencesUniversity at Buffalo,140 Farber Hall, 3435 Main Street,Buffalo, NY 14214, USA
| | - Dongwook Kim
- Biochemistry Department,School of Medicine and Biomedical SciencesUniversity at Buffalo,140 Farber Hall, 3435 Main Street,Buffalo, NY 14214, USA
| | - Trivikram Rajkhowa
- Biochemistry Department,School of Medicine and Biomedical SciencesUniversity at Buffalo,140 Farber Hall, 3435 Main Street,Buffalo, NY 14214, USA
| | - Facundo Cutuli
- Biochemistry Department,School of Medicine and Biomedical SciencesUniversity at Buffalo,140 Farber Hall, 3435 Main Street,Buffalo, NY 14214, USA
| |
Collapse
|
26
|
Di Ciano LA, Azurmendi PJ, Colombero C, Levin G, Oddo EM, Arrizurieta EE, Nowicki S, Ibarra FR. Defective renal dopamine function and sodium-sensitive hypertension in adult ovariectomized Wistar rats: role of the cytochrome P-450 pathway. Am J Physiol Renal Physiol 2015; 308:F1358-68. [PMID: 25925257 DOI: 10.1152/ajprenal.00450.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 04/28/2015] [Indexed: 01/11/2023] Open
Abstract
We have previously shown that ovariectomy in adult Wistar rats under normal sodium (NS) intake results in an overexpression of the total Na(+)-K(+)-ATPase (NKA) α1-subunit (Di Ciano LA, Azurmendi PJ, Toledo JE, Oddo EM, Zotta E, Ochoa F, Arrizurieta EE, Ibarra FR. Clin Exp Hypertens 35: 475-483, 2013). Upon high sodium (HS) intake, ovariectomized (oVx) rats developed defective NKA phosphorylation, a decrease in sodium excretion, and an increment in mean blood pressure (MBP). Since NKA phosphorylation is modulated by dopamine (DA), the aim of this study was to compare the intracellular response of the renal DA system leading to NKA phosphorylation upon sodium challenge in intact female (IF) and oVx rats. In IF rats, HS caused an increase in urinary DA and sodium, in NKA phosphorylation state, in cytochrome P-4504A (CYP4A) expression, and in 20-HETE production, while MBP kept normal. Blockade of the D1 receptor (D1R) with the D1-like receptor antagonist SCH 23390 in IFHS rats shifted NKA into a more dephosphorylated state, decreased sodium excretion by 50%, and increased MBP. In oVxNS rats, D1R expression was reduced and D3R expression was increased, and under HS intake sodium excretion was lower and MBP higher than in IFHS rats (both P < 0.05), NKA was more dephosphorylated than in IFHS, and CYP4A expression or 20-HETE production did not change. Blockade of D1R in oVxHS rats changed neither NKA phosphorylation state nor sodium excretion or MBP. D2R and PKCα expression did not vary among groups. The alteration of the renal DA system produced by ovariectomy could account for the defective NKA phosphorylation, the inefficient excretion of sodium load, and the development of salt-sensitive hypertension.
Collapse
Affiliation(s)
- Luis A Di Ciano
- Instituto de Investigaciones Médicas A Lanari, Buenos Aires University, Buenos Aires, Argentina
| | - Pablo J Azurmendi
- Instituto de Investigaciones Médicas A Lanari, Buenos Aires University, Buenos Aires, Argentina
| | - Cecilia Colombero
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Gloria Levin
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Elisabet M Oddo
- Instituto de Investigaciones Médicas A Lanari, Buenos Aires University, Buenos Aires, Argentina
| | - Elvira E Arrizurieta
- Instituto de Investigaciones Médicas A Lanari, Buenos Aires University, Buenos Aires, Argentina; CONICET, Buenos Aires, Argentina
| | - Susana Nowicki
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá" (CEDIE), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-FEI-División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Buenos Aires, Argentina
| | - Fernando R Ibarra
- Instituto de Investigaciones Médicas A Lanari, Buenos Aires University, Buenos Aires, Argentina; Departamento de Fisiología, Facultad de Medicina, Buenos Aires University, Buenos Aires, Argentina; and
| |
Collapse
|
27
|
Cuevas S, Yang Y, Konkalmatt P, Asico LD, Feranil J, Jones J, Villar VA, Armando I, Jose PA. Role of nuclear factor erythroid 2-related factor 2 in the oxidative stress-dependent hypertension associated with the depletion of DJ-1. Hypertension 2015; 65:1251-7. [PMID: 25895590 DOI: 10.1161/hypertensionaha.114.04525] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 03/26/2015] [Indexed: 01/11/2023]
Abstract
Renal dopamine 2 receptor dysfunction is associated with oxidative stress and high blood pressure (BP). We have reported that DJ-1, an oxidative stress response protein, is positively regulated by dopamine 2 receptor in the kidney. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates the expression of several antioxidant genes. We tested the hypothesis that Nrf2 is involved in the renal DJ-1-mediated inhibition of reactive oxygen species production. We have reported that silencing dopamine 2 receptor in mouse renal proximal tubule cells decreases the expression of DJ-1. We now report that silencing DJ-1 or dopamine 2 receptor in mouse proximal tubule cells and mouse kidneys decreases Nrf2 expression and activity and increases reactive oxygen species production; BP is also increased in mice in which renal DJ-1 or dopamine 2 receptor is silenced. DJ-1(-/-) mice have decreased renal Nrf2 expression and activity and increased nitro-tyrosine levels and BP. Silencing Nrf2 in mouse proximal tubule cells does not alter the expression of DJ-1 or dopamine 2 receptor, indicating that Nrf2 is downstream of dopamine 2 receptor and DJ-1. An Nrf2 inducer, bardoxolone, normalizes the systolic BP and renal malondialdehyde levels in DJ-1(-/-) mice without affecting them in their wild-type littermates. Because Nrf2 ubiquitination is increased in DJ-1(-/-) mice, we conclude that the protective effect of DJ-1 on renal oxidative stress is mediated, in part, by preventing Nrf2 degradation. Moreover, renal dopamine 2 receptor and DJ-1 are necessary for normal Nrf2 activity to keep a normal redox balance and BP.
Collapse
Affiliation(s)
- Santiago Cuevas
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore.
| | - Yu Yang
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| | - Prasad Konkalmatt
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| | - Laureano D Asico
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| | - Jun Feranil
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| | - John Jones
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| | - Van Anthony Villar
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| | - Ines Armando
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| | - Pedro A Jose
- From the Department of Medicine, Division of Nephrology (S.C., Y.Y., P.K., L.D.A., J.F., J.J., V.A.V., I.A., P.A.J.) and Department of Physiology (P.A.J.), University of Maryland School of Medicine, Baltimore
| |
Collapse
|
28
|
Wang Y, Wang D, Chu C, Mu JJ, Wang M, Liu FQ, Xie BQ, Yang F, Dong ZZ, Yuan ZY. Effect of Salt Intake and Potassium Supplementation on Urinary Renalase and Serum Dopamine Levels in Chinese Adults. Cardiology 2015; 130:242-8. [DOI: 10.1159/000371794] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/23/2014] [Indexed: 11/19/2022]
Abstract
Objective: The aim of our study was to assess the effects of altered salt and potassium intake on urinary renalase and serum dopamine levels in humans. Methods: Forty-two subjects (28-65 years of age) were selected from a rural community of northern China. All subjects were sequentially maintained on a low-salt diet for 7 days (3.0 g/day of NaCl), a high-salt diet for an additional 7 days (18.0 g/day of NaCl), and a high-salt diet with potassium supplementation for a final 7 days (18.0 g/day of NaCl + 4.5 g/day of KCl). Results: Urinary renalase excretions were significantly higher during the high-salt diet intervention than during the low-salt diet. During high-potassium intake, urinary renalase excretions were not significantly different from the high-salt diet, whereas they were significantly higher than the low-salt levels. Serum dopamine levels exhibited similar trends across the interventions. Additionally, a significant positive relationship was observed between the urine renalase and serum dopamine among the different dietary interventions. Also, 24-hour urinary sodium excretion positively correlated with urine renalase and serum dopamine in the whole population. Conclusions: The present study indicates that dietary salt intake and potassium supplementation increase urinary renalase and serum dopamine levels in Chinese subjects.
Collapse
|
29
|
The importance of G protein-coupled receptor kinase 4 (GRK4) in pathogenesis of salt sensitivity, salt sensitive hypertension and response to antihypertensive treatment. Int J Mol Sci 2015; 16:5741-9. [PMID: 25775155 PMCID: PMC4394502 DOI: 10.3390/ijms16035741] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/06/2015] [Accepted: 01/15/2015] [Indexed: 12/28/2022] Open
Abstract
Salt sensitivity is probably caused by either a hereditary or acquired defect of salt excretion by the kidney, and it is reasonable to consider that this is the basis for differences in hypertension between black and white people. Dopamine acts in an autocrine/paracrine fashion to promote natriuresis in the proximal tubule and thick ascending loop of Henle. G-protein receptor kinases (or GRKs) are serine and threonine kinases that phosphorylate G protein-coupled receptors in response to agonist stimulation and uncouple the dopamine receptor from its G protein. This results in a desensitisation process that protects the cell from repeated agonist exposure. GRK4 activity is increased in spontaneously hypertensive rats, and infusion of GRK4 antisense oligonucleotides attenuates the increase in blood pressure (BP). This functional defect is replicated in the proximal tubule by expression of GRK4 variants namely p.Arg65Leu, p.Ala142Val and p.Val486Ala, in cell lines, with the p.Ala142Val showing the most activity. In humans, GRK4 polymorphisms were shown to be associated with essential hypertension in Australia, BP regulation in young adults, low renin hypertension in Japan and impaired stress-induced Na excretion in normotensive black men. In South Africa, GRK4 polymorphisms are more common in people of African descent, associated with impaired Na excretion in normotensive African people, and predict blood pressure response to Na restriction in African patients with mild to moderate essential hypertension. The therapeutic importance of the GRK4 single nucleotide polymorphisms (SNPs) was emphasised in the African American Study of Kidney Disease (AASK) where African-Americans with hypertensive nephrosclerosis were randomised to receive amlodipine, ramipril or metoprolol. Men with the p.Ala142Val genotype were less likely to respond to metoprolol, especially if they also had the p.Arg65Leu variant. Furthermore, in the analysis of response to treatment in two major hypertension studies, the 65Leu/142Val heterozygote predicted a significantly decreased response to atenolol treatment, and the 65Leu/142Val heterozygote and 486Val homozygote were associated in an additive fashion with adverse cardiovascular outcomes, independent of BP. In conclusion, there is considerable evidence that GRK4 variants are linked to impaired Na excretion, hypertension in animal models and humans, therapeutic response to dietary Na restriction and response to antihypertensive drugs. It may also underlie the difference in hypertension between different geographically derived population groups, and form a basis for pharmacogenomic approaches to treatment of hypertension.
Collapse
|
30
|
Li Y, Wang N, Chen C, He D, Yang J, Zeng C. Inhibitory effect of D3dopamine receptor on migration of vascular smooth muscle cells induced by synergistic effect of angiotensin II and aldosterone. Clin Exp Hypertens 2014; 37:288-93. [DOI: 10.3109/10641963.2014.960971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
31
|
Arystarkhova E, Ralph DL, Liu YB, Bouley R, McDonough AA, Sweadner KJ. Paradoxical activation of the sodium chloride cotransporter (NCC) without hypertension in kidney deficient in a regulatory subunit of Na,K-ATPase, FXYD2. Physiol Rep 2014; 2:2/12/e12226. [PMID: 25472608 PMCID: PMC4332208 DOI: 10.14814/phy2.12226] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Na,K‐ATPase generates the driving force for sodium reabsorption in the kidney.
Na,K‐ATPase functional properties are regulated by small proteins belonging to the FXYD
family. In kidney FXYD2 is the most abundant: it is an inhibitory subunit expressed in almost every
nephron segment. Its absence should increase sodium pump activity and promote Na+
retention, however, no obvious renal phenotype was detected in mice with global deletion of FXYD2
(Arystarkhova et al. 2013). Here, increased total cortical Na,K‐ATPase activity was
documented in the Fxyd2−/− mouse, without increased
α1β1 subunit expression. We tested the hypothesis
that adaptations occur in distal convoluted tubule (DCT), a major site of sodium adjustments.
Na,K‐ATPase immunoreactivity in DCT was unchanged, and there was no DCT hypoplasia. There was
a marked activation of thiazide‐sensitive sodium chloride cotransporter (NCC; Slc12a3) in
DCT, predicted to increase Na+ reabsorption in this segment. Specifically, NCC
total increased 30% and NCC phosphorylated at T53 and S71, associated with activation,
increased 4‐6 fold. The phosphorylation of the closely related thick ascending limb (TAL)
apical NKCC2 (Slc12a1) increased at least twofold. Abundance of the total and cleaved (activated)
forms of ENaC α‐subunit was not different between genotypes.
Nonetheless, no elevation of blood pressure was evident despite the fact that NCC and NKCC2 are in
states permissive for Na+ retention. Activation of NCC and NKCC2 may reflect an
intracellular linkage to elevated Na,K‐ATPase activity or a compensatory response to
Na+ loss proximal to the TAL and DCT. We discovered a substantial activation of renal NCC cotransporter in mice genetically depleted
for the regulatory inhibitory subunit of Na,K‐ATPase, FXYD2. Surprisingly, no significant
changes in urine output as well as elevation of blood pressure were detected suggesting compensatory
adaptation elsewhere in nephron
Collapse
Affiliation(s)
- Elena Arystarkhova
- Laboratory of Membrane Biology, Massachusetts General Hospital, Boston, Massachusetts
| | - Donna L Ralph
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yi Bessie Liu
- Laboratory of Membrane Biology, Massachusetts General Hospital, Boston, Massachusetts
| | - Richard Bouley
- MGH Center for Systems Biology, Program in Membrane Biology, Massachusetts General Hospital, Boston, Massachusetts
| | - Alicia A McDonough
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Kathleen J Sweadner
- Laboratory of Membrane Biology, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
32
|
Quelhas-Santos J, Serrão MP, Soares-Silva I, Fernandes-Cerqueira C, Simões-Silva L, Pinho MJ, Remião F, Sampaio-Maia B, Desir GV, Pestana M. Renalase regulates peripheral and central dopaminergic activities. Am J Physiol Renal Physiol 2014; 308:F84-91. [PMID: 25411385 DOI: 10.1152/ajprenal.00274.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Renalase is a recently identified FAD/NADH-dependent amine oxidase mainly expressed in kidney that is secreted into blood and urine where it was suggested to metabolize catecholamines. The present study evaluated central and peripheral dopaminergic activities in the renalase knockout (KO) mouse model and examined the changes induced by recombinant renalase (RR) administration on plasma and urine catecholamine levels. Compared with wild-type (WT) mice, KO mice presented increased plasma levels of epinephrine (Epi), norepinephrine (NE), and dopamine (DA) that were accompanied by increases in the urinary excretion of Epi, NE, DA. In addition, the KO mice presented an increase in urinary DA-to-l-3,4-dihydroxyphenylalanine (l-DOPA) ratios without changes in renal tubular aromatic-l-amino acid decarboxylase (AADC) activity. By contrast, the in vivo administration of RR (1.5 mg/kg sc) to KO mice was accompanied by significant decreases in plasma levels of Epi, DA, and l-DOPA as well as in urinary excretion of Epi, DA, and DA-to-l-DOPA ratios notwithstanding the accompanied increase in renal AADC activity. In addition, the increase in renal DA output observed in renalase KO mice was accompanied by an increase in the expression of the L-type amino acid transporter like (LAT) 1 that is reversed by the administration of RR in these animals. These results suggest that the overexpression of LAT1 in the renal cortex of the renalase KO mice might contribute to the enhanced l-DOPA availability/uptake and consequently to the activation of the renal dopaminergic system in the presence of renalase deficiency.
Collapse
Affiliation(s)
- Janete Quelhas-Santos
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal; Nephrology and Infectious Diseases Research and Development Group, Instituto Nacional de Engenharia Biomédica-(I3S);
| | - Maria Paula Serrão
- Faculdade de Medicina da Universidade do Porto, Department of Pharmacology and Therapeutics, Porto, Portugal
| | - Isabel Soares-Silva
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal; Nephrology and Infectious Diseases Research and Development Group, Instituto Nacional de Engenharia Biomédica-(I3S)
| | | | - Liliana Simões-Silva
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Maria João Pinho
- Faculdade de Medicina da Universidade do Porto, Department of Pharmacology and Therapeutics, Porto, Portugal
| | - Fernando Remião
- Centro de Química da Universidade do Porto/Serviço de Toxicologia, Faculdade de Farmácia, University of Porto, Porto, Portugal
| | - Benedita Sampaio-Maia
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal; Faculty of Dental Medicine, University of Porto, Porto, Portugal
| | - Gary V Desir
- Department of Medicine, Veterans Affairs Connecticut Healthcree System, Yale University, New Haven, Connecticut
| | - Manuel Pestana
- Nephrology and Infectious Diseases Research and Development Group, Instituto Nacional de Engenharia Biomédica-(I3S); Faculdade de Medicina da Universidade do Porto, Department of Renal, Urological, and Infectious Diseases, Porto, Portugal; and Department of Nephrology, São João Hospital Center, Entidade Pública Empresarial, Porto, Portugal
| |
Collapse
|
33
|
Signaling pathways involved in renal oxidative injury: role of the vasoactive peptides and the renal dopaminergic system. JOURNAL OF SIGNAL TRANSDUCTION 2014; 2014:731350. [PMID: 25436148 PMCID: PMC4243602 DOI: 10.1155/2014/731350] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/16/2014] [Indexed: 12/24/2022]
Abstract
The physiological hydroelectrolytic balance and the redox steady state in the kidney are accomplished by an intricate interaction between signals from extrarenal and intrarenal sources and between antinatriuretic and natriuretic factors. Angiotensin II, atrial natriuretic peptide and intrarenal dopamine play a pivotal role in this interactive network. The balance between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide, by one side, and the prooxidant effect of the renin angiotensin system, by the other side, contributes to ensuring the normal function of the kidney. Different pathological scenarios, as nephrotic syndrome and hypertension, where renal sodium excretion is altered, are associated with an impaired interaction between two natriuretic systems as the renal dopaminergic system and atrial natriuretic peptide that may be involved in the pathogenesis of renal diseases. The aim of this review is to update and comment the most recent evidences about the intracellular pathways involved in the relationship between endogenous antioxidant agents like the renal dopaminergic system and atrial natriuretic peptide and the prooxidant effect of the renin angiotensin system in the pathogenesis of renal inflammation.
Collapse
|
34
|
Ennis RC, Asico LD, Armando I, Yang J, Feranil JB, Jurgens JA, Escano CS, Yu P, Wang X, Sibley DR, Jose PA, Villar VAM. Dopamine D₁-like receptors regulate the α₁A-adrenergic receptor in human renal proximal tubule cells and D₁-like dopamine receptor knockout mice. Am J Physiol Renal Physiol 2014; 307:F1238-48. [PMID: 25339698 DOI: 10.1152/ajprenal.00119.2014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The homeostatic control of blood pressure hinges upon the delicate balance between prohypertensinogenic and antihypertensinogenic systems. D₁-like dopamine receptors [dopamine D₁ and D₅ receptors (D₁Rs and D₅Rs, respectively)] and the α₁A-adrenergic receptor (α₁A-AR) are expressed in the renal proximal tubule and engender opposing effects on Na(+) transport, i.e., natriuresis (via D₁Rs and D5Rs) or antinatriuresis (via α₁A-ARs). We tested the hypothesis that the D₁R/D₅R regulates the α₁A-AR. D₁-like dopamine receptors coimmunoprecipitated, colocalized, and cofractionated with α₁A-ARs in lipid rafts in immortalized human renal proximal tubule cells. Long-term treatment with the D₁R/D₅R agonist fenoldopam resulted in decreased D₁R and D₅R expression but increased α₁A-AR abundance in the plasma membrane. Short-term fenoldopam treatment stimulated the translocation of Na(+)-K(+)-ATPase from the plasma membrane to the cytosol that was partially reversed by an α₁A-AR agonist, which by itself induced Na(+)-K(+)-ATPase translocation from the cytosol to the plasma membrane. The α₁A-AR-specific agonist A610603 also minimized the ability of fenoldopam to inhibit Na(+)-K(+)-ATPase activity. To determine the interaction among D₁Rs, D₅Rs, and α₁A-ARs in vivo, we used phenylephrine and A610603 to decrease Na(+) excretion in several D1-like dopamine receptor knockout mouse strains. Phenylephrine and A61603 treatment resulted in a partial reduction of urinary Na(+) excretion in wild-type mice and its abolition in D1R knockout, D₅R knockout, and D₁R-D₅R double-knockout mice. Our results demonstrate the ability of the D₁-like dopamine receptors to regulate the expression and activity of α₁A-AR. Elucidating the intricacies of the interaction among these receptors is crucial for a better understanding of the crosstalk between anti- and pro-hypertensive systems.
Collapse
Affiliation(s)
- Riley Charles Ennis
- Thomas Jefferson High School for Science and Technology, Alexandria, Virgina
| | - Laureano D Asico
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ines Armando
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jian Yang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jun B Feranil
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Julie A Jurgens
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Crisanto S Escano
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peiying Yu
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Xiaoyan Wang
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Pedro A Jose
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Van Anthony M Villar
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland;
| |
Collapse
|
35
|
Atrial natriuretic peptide and renal dopaminergic system: a positive friendly relationship? BIOMED RESEARCH INTERNATIONAL 2014; 2014:710781. [PMID: 25013796 PMCID: PMC4075025 DOI: 10.1155/2014/710781] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/12/2014] [Accepted: 05/12/2014] [Indexed: 01/11/2023]
Abstract
Sodium metabolism by the kidney is accomplished by an intricate interaction between signals from extrarenal and intrarenal sources and between antinatriuretic and natriuretic factors. Renal dopamine plays a central role in this interactive network. The natriuretic hormones, such as the atrial natriuretic peptide, mediate some of their effects by affecting the renal dopaminergic system. Renal dopaminergic tonus can be modulated at different steps of dopamine metabolism (synthesis, uptake, release, catabolism, and receptor sensitization) which can be regulated by the atrial natriuretic peptide. At tubular level, dopamine and atrial natriuretic peptide act together in a concerted manner to promote sodium excretion, especially through the overinhibition of Na+, K+-ATPase activity. In this way, different pathological scenarios where renal sodium excretion is dysregulated, as in nephrotic syndrome or hypertension, are associated with impaired action of renal dopamine and/or atrial natriuretic peptide, or as a result of impaired interaction between these two natriuretic systems. The aim of this review is to update and comment on the most recent evidences demonstrating how the renal dopaminergic system interacts with atrial natriuretic peptide to control renal physiology and blood pressure through different regulatory pathways.
Collapse
|
36
|
Colombero C, Venara M, Gonzalez D, Roman RJ, Nowicki S. Cytochrome P4504A inhibitors attenuate the exaggerated natriuretic response to volume expansion in thyroidectomized rats. Physiol Rep 2014; 2:2/6/e12040. [PMID: 24920124 PMCID: PMC4208633 DOI: 10.14814/phy2.12040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Cecilia Colombero
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE); CONICET - FEI - División de Endocrinología; Hospital de Niños Ricardo Gutiérrez; Buenos Aires Argentina
| | - Marcela Venara
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE); CONICET - FEI - División de Endocrinología; Hospital de Niños Ricardo Gutiérrez; Buenos Aires Argentina
| | - Daniel Gonzalez
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE); CONICET - FEI - División de Endocrinología; Hospital de Niños Ricardo Gutiérrez; Buenos Aires Argentina
| | - Richard J. Roman
- Department of Pharmacology and Toxicology; The University of Mississippi Medical Center; Jackson Mississippi
| | - Susana Nowicki
- Centro de Investigaciones Endocrinológicas “Dr. César Bergadá” (CEDIE); CONICET - FEI - División de Endocrinología; Hospital de Niños Ricardo Gutiérrez; Buenos Aires Argentina
| |
Collapse
|
37
|
Granda ML, Schroeder FA, Borra RHJ, Schauer N, Aisaborhale E, Guimaraes AR, Hooker JM. First D1-like receptor PET imaging of the rat and primate kidney: implications for human disease monitoring. Am J Physiol Renal Physiol 2014; 307:F116-21. [PMID: 24808534 DOI: 10.1152/ajprenal.00111.2014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The intrarenal dopamine system is important for signaling and natriuresis, and significant dysfunction is associated with hypertension and kidney disease in ex vivo studies. Dopamine receptors also modulate and are modulated by the renin-angiotensin-aldosterone system. Here, we show the first in vivo measurement of D1-like receptors in the renal cortex of Sprague-Dawley rat and Papio anubis baboon using [(11)C]NNC 112, a positron emission tomography radioligand for D1-like receptors. In addition, we show a D1-like binding potential response to angiotensin II blockade in rats using losartan. Demonstration of self-saturable binding in the rat as well as specific and saturable binding in Papio anubis validate the use of [(11)C]NNC 112 in the first in vivo measurement of renal dopamine D1-like receptors. Furthermore, [(11)C]NNC 112 is a radioligand tool already validated for use in probing human central nervous system (CNS) D1-like receptors. Our work demonstrates specific and saturable non-CNS binding in higher animals and the ability to quantify physiological response to drug treatment and provides a clear path to extend use of [(11)C]NNC 112 to study renal dopamine in humans.
Collapse
Affiliation(s)
- Michael L Granda
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and Eastern Virginia Medical School, Norfolk, Virginia
| | - Frederick A Schroeder
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| | - Ronald H J Borra
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| | - Nathan Schauer
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| | - Ehimen Aisaborhale
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| | - Alexander R Guimaraes
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts; and
| |
Collapse
|
38
|
Zhang MZ, Wang Y, Yao B, Gewin L, Wei S, Capdevila JH, Harris RC. Role of epoxyeicosatrienoic acids (EETs) in mediation of dopamine's effects in the kidney. Am J Physiol Renal Physiol 2013; 305:F1680-6. [PMID: 24154693 DOI: 10.1152/ajprenal.00409.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
We have recently demonstrated that intrarenal dopamine plays an important role in preventing the development of systemic hypertension. Similarly, renal cytochrome P-450 (CYP)-epoxygenase-derived arachidonic acid metabolites, epoxyeicosatrienoic acids (EETs), also are antihypertensive through inhibiting sodium reabsorption and vasodilation. The potential interaction between renal dopamine and epoxygenase systems was investigated. Catechol-O-methyl-transferase (COMT)(-/-) mice with increased intrarenal dopamine levels and proximal tubule deletion of aromatic amino acid decarboxylase (ptAADC(-/-)) mice with renal dopamine deficiency were treated with a low-salt diet or high-salt diet for 2 wk. Wild-type or Cyp2c44(-/-) mice were treated with gludopa, which selectively increased renal dopamine levels. In low salt-treated mice, urinary EET levels were related to renal dopamine levels, being highest in COMT(-/-) mice and lowest in ptAADC(-/-) mice. In high salt-treated mice, total EET and individual EET levels in both the kidney and urine were also highest in COMT(-/-) mice and lowest in ptAADC(-/-) mice. Selective increases in renal dopamine in response to gludopa administration led to marked increases in both total and all individual EET levels in the kidney without any changes in blood levels. qRT-PCR and immunoblotting indicated that gludopa increased renal Cyp2c44 mRNA and protein levels. Gludopa induced marked increases in urine volume and urinary sodium excretion in wild-type mice. In contrast, gludopa did not induce significant increases in urine volume or urinary sodium excretion in Cyp2c44(-/-) mice. These studies demonstrate that renal EET levels are maintained by intrarenal dopamine, and Cyp2c44-derived EETs play an important role in intrarenal dopamine-induced natriuresis and diuresis.
Collapse
Affiliation(s)
- Ming-Zhi Zhang
- Div. of Nephrology, C3121 MCN, Vanderbilt Univ. School of Medicine and Nashville Veterans Affairs Hospital, Nashville, TN 37232.
| | | | | | | | | | | | | |
Collapse
|
39
|
Quelhas-Santos J, Sampaio-Maia B, Simões-Silva L, Serrão P, Fernandes-Cerqueira C, Soares-Silva I, Pestana M. Sodium-dependent modulation of systemic and urinary renalase expression and activity in the rat remnant kidney. J Hypertens 2013; 31:543-52; discussion 552-3. [PMID: 23314744 DOI: 10.1097/hjh.0b013e32835d6e34] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The present study examined the influence of high-sodium intake on systemic and urinary renalase levels and activity in 3/4 nephrectomized (3/4nx) and Sham rats. RESULTS The reduced circulating renalase levels in 3/4nx rats during normal-sodium intake were accompanied by increased plasma renalase activity. The sodium-induced increase of blood pressure in 3/4nx rats was accompanied by significant decreases in circulating renalase levels and activity as well as by a significant decrease in cardiac renalase levels in 3/4nx rats but not in Sham rats. During normal-sodium intake, no significant differences were observed in either urine renalase levels or activity between 3/4nx and Sham rats, not withstanding the ∼75% decrease in daily urine dopamine output observed in the rat remnant kidney. During high-sodium intake, urinary renalase levels increased in both 3/4nx and Sham groups by three-fold whereas urinary renalase activity increased in 3/4nx and Sham rats by greater than twelve-fold and greater than four-fold, respectively. This was accompanied by sodium-induced increases in daily urinary dopamine output in both 3/4nx and Sham rats by ∼2.3-fold and ∼1.6-fold, respectively. CONCLUSION The reduced circulating renalase levels in 3/4nx rats are accompanied by increased plasma renalase activity, which appears to be related with decreased inhibition of the circulating enzyme. Differences in systemic and urinary renalase levels and activity between 3/4nx and Sham rats during high-sodium intake may contribute to activation of the sympathetic nervous system, hypertension and enhanced cardiovascular risk in CKD but do not appear to account for the decrease in renal dopaminergic activity in the rat remnant kidney.
Collapse
Affiliation(s)
- Janete Quelhas-Santos
- Nephrology Research and Development Unit, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | | | | | | | | | | |
Collapse
|
40
|
Koepsell H. The SLC22 family with transporters of organic cations, anions and zwitterions. Mol Aspects Med 2013; 34:413-35. [PMID: 23506881 DOI: 10.1016/j.mam.2012.10.010] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 08/18/2012] [Indexed: 12/14/2022]
Abstract
The SLC22 family contains 13 functionally characterized human plasma membrane proteins each with 12 predicted α-helical transmembrane domains. The family comprises organic cation transporters (OCTs), organic zwitterion/cation transporters (OCTNs), and organic anion transporters (OATs). The transporters operate as (1) uniporters which mediate facilitated diffusion (OCTs, OCTNs), (2) anion exchangers (OATs), and (3) Na(+)/zwitterion cotransporters (OCTNs). They participate in small intestinal absorption and hepatic and renal excretion of drugs, xenobiotics and endogenous compounds and perform homeostatic functions in brain and heart. Important endogeneous substrates include monoamine neurotransmitters, l-carnitine, α-ketoglutarate, cAMP, cGMP, prostaglandins, and urate. It has been shown that mutations of the SLC22 genes encoding these transporters cause specific diseases like primary systemic carnitine deficiency and idiopathic renal hypouricemia and are correlated with diseases such as Crohn's disease and gout. Drug-drug interactions at individual transporters may change pharmacokinetics and toxicities of drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, 97070 Würzburg, Germany.
| |
Collapse
|
41
|
Concerted action of ANP and dopamine D1-receptor to regulate sodium homeostasis in nephrotic syndrome. BIOMED RESEARCH INTERNATIONAL 2013; 2013:397391. [PMID: 23956981 PMCID: PMC3727124 DOI: 10.1155/2013/397391] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/23/2013] [Accepted: 06/24/2013] [Indexed: 12/22/2022]
Abstract
The edema formation in nephrotic syndrome (NS) is associated with a blunted response to atrial natriuretic peptide (ANP). The natriuretic effects of ANP have been related to renal dopamine D1-receptors (D1R). We examined the interaction between ANP and renal D1R in rats with puromycin aminonucleoside-induced NS (PAN-NS). Urinary sodium, cyclic guanosine monophosphate (cGMP) excretion, and D1R protein expression and localization in renal tubules were evaluated in PAN-NS and control rats before and during volume expansion (VE). The effects of zaprinast (phosphodiesterase type 5 inhibitor), alone or in combination with Sch-23390 (D1R antagonist), were examined in both groups. The increased natriuresis and urinary cGMP excretion evoked by acute VE were blunted in PAN-NS despite increased levels of circulating ANP. This was accompanied in PAN-NS by a marked decrease of D1R expression in the renal tubules. Infusion of zaprinast in PAN-NS resulted in increased urinary excretion of cGMP and sodium to similar levels of control rats and increased expression of D1R in the plasma membrane of renal tubular cells. Combined administration of Sch-23390 and zaprinast prevented natriuresis and increased cGMP excretion induced by zaprinast alone. We conclude that D1R may play a major role in the ANP resistance observed in PAN-NS.
Collapse
|
42
|
Abstract
The kidney plays a fundamental role in maintaining body salt and fluid balance and blood pressure homeostasis through the actions of its proximal and distal tubular segments of nephrons. However, proximal tubules are well recognized to exert a more prominent role than distal counterparts. Proximal tubules are responsible for reabsorbing approximately 65% of filtered load and most, if not all, of filtered amino acids, glucose, solutes, and low molecular weight proteins. Proximal tubules also play a key role in regulating acid-base balance by reabsorbing approximately 80% of filtered bicarbonate. The purpose of this review article is to provide a comprehensive overview of new insights and perspectives into current understanding of proximal tubules of nephrons, with an emphasis on the ultrastructure, molecular biology, cellular and integrative physiology, and the underlying signaling transduction mechanisms. The review is divided into three closely related sections. The first section focuses on the classification of nephrons and recent perspectives on the potential role of nephron numbers in human health and diseases. The second section reviews recent research on the structural and biochemical basis of proximal tubular function. The final section provides a comprehensive overview of new insights and perspectives in the physiological regulation of proximal tubular transport by vasoactive hormones. In the latter section, attention is particularly paid to new insights and perspectives learnt from recent cloning of transporters, development of transgenic animals with knockout or knockin of a particular gene of interest, and mapping of signaling pathways using microarrays and/or physiological proteomic approaches.
Collapse
Affiliation(s)
- Jia L Zhuo
- Laboratory of Receptor and Signal Transduction, Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| | | |
Collapse
|
43
|
Hu MC, Di Sole F, Zhang J, McLeroy P, Moe OW. Chronic regulation of the renal Na(+)/H(+) exchanger NHE3 by dopamine: translational and posttranslational mechanisms. Am J Physiol Renal Physiol 2013; 304:F1169-80. [PMID: 23427139 DOI: 10.1152/ajprenal.00630.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The intrarenal autocrine/paracrine dopamine (DA) system contributes to natriuresis in response to both acute and chronic Na(+) loads. While the acute DA effect is well described, how DA induces natriuresis chronically is not known. We used an animal and a cell culture model to study the chronic effect of DA on a principal renal Na(+) transporter, Na(+)/H(+) exchanger-3 (NHE3). Intraperitoneal injection of Gludopa in rats for 2 days elevated DA excretion and decreased total renal cortical and apical brush-border NHE3 antigen. Chronic treatment of an opossum renal proximal cell line with DA decreased NHE3 activity, cell surface and total cellular NHE3 antigen, but not NHE3 transcript. The decrease in NHE3 antigen was dose and time dependent with maximal inhibition at 16-24 h and half maximal effect at 3 × 10(-7) M. This is in contradistinction to the acute effect of DA on NHE3 (half maximal at 2 × 10(-6) M), which was not associated with changes in total cellular NHE3 protein. The DA-induced decrease in total NHE3 protein was associated with decrease in NHE3 translation and mediated by cis-sequences in the NHE3 5'-untranslated region. DA also decreased cell surface and total cellular NHE3 protein half-life. The DA-induced decrease in total cellular NHE3 was partially blocked by proteasome inhibition but not by lysosome inhibition, and DA increased ubiquitylation of total and surface NHE3. In summary, chronic DA inhibits NHE3 with mechanisms distinct from its acute action and involves decreased NHE3 translation and increased NHE3 degradation, which are novel mechanisms for NHE3 regulation.
Collapse
Affiliation(s)
- Ming Chang Hu
- Dept. of Internal Medicine, Univ. of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8885, USA
| | | | | | | | | |
Collapse
|
44
|
Villar VAM, Armando I, Sanada H, Frazer LC, Russo CM, Notario PM, Lee H, Comisky L, Russell HA, Yang Y, Jurgens JA, Jose PA, Jones JE. Novel role of sorting nexin 5 in renal D(1) dopamine receptor trafficking and function: implications for hypertension. FASEB J 2012. [PMID: 23195037 DOI: 10.1096/fj.12-208439] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The D1 dopamine receptor (D1R) is widely expressed in the kidney and plays a crucial role in blood pressure regulation. Although much is known about D1R desensitization, especially through G-protein-coupled receptor kinase 4 (GRK4), comparatively little is known about other aspects of D1R trafficking and the proteins involved in the process. We now report the discovery of a dynamic interaction between sorting nexin 5 (SNX5), a component of the mammalian retromer, and D1R in human renal epithelial cells. We show that internalization of agonist-activated D1R is regulated by both SNX5 and GRK4, and that SNX5 is critical to the recycling of the receptor to the plasma membrane. SNX5 depletion increases agonist-activated D1R phosphorylation (>50% at basal condition), prevents D1R internalization and cAMP response, and delays receptor recycling compared to mock siRNA-transfected controls. Moreover, renal restricted subcapsular infusion of Snx5-specific siRNA (vs. mock siRNA) decreases sodium excretion (Δ=-0.2±0.005 mEq/mg creatinine) and further elevates the systolic blood pressure (Δ=48±5 mm Hg) in spontaneously hypertensive rats, indicating that SNX5 depletion impairs renal D1R function. These studies demonstrate an essential role for SNX5 in regulating D1R function, which may have important diagnostic, prognostic, and therapeutic implications in the management of essential hypertension.
Collapse
Affiliation(s)
- Van Anthony M Villar
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, 20 Penn St., Ste. S003C, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chugh G, Pokkunuri I, Asghar M. Renal dopamine and angiotensin II receptor signaling in age-related hypertension. Am J Physiol Renal Physiol 2012; 304:F1-7. [PMID: 23097467 DOI: 10.1152/ajprenal.00441.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Kidneys play a vital role in long-term regulation of blood pressure. This is achieved by actions of many renal and nonrenal factors acting on the kidney that help maintain the body's water and electrolyte balance and thus control blood pressure. Several endogenously formed or circulating hormones/peptides, by acting within the kidney, regulate fluid and water homeostasis and blood pressure. Dopamine and angiotensin II are the two key renal factors that, via acting on their receptors and counterregulating each other's function, maintain water and sodium balance. In this review, we provide recent advances in the signaling cascades of these renal receptors, especially at the level of their cross talk, and discuss their roles in blood pressure regulation in the aging process.
Collapse
Affiliation(s)
- Gaurav Chugh
- Heart and Kidney Institute, College of Pharmacy, Univ. of Houston, Houston, TX 77204, USA
| | | | | |
Collapse
|
46
|
Zhang MZ, Yao B, Yang S, Yang H, Wang S, Fan X, Yin H, Fogo AB, Moeckel GW, Harris RC. Intrarenal dopamine inhibits progression of diabetic nephropathy. Diabetes 2012; 61:2575-84. [PMID: 22688335 PMCID: PMC3447896 DOI: 10.2337/db12-0046] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The kidney has a local intrarenal dopaminergic system, and in the kidney, dopamine modulates renal hemodynamics, inhibits salt and fluid reabsorption, antagonizes the renin-angiotensin system, and inhibits oxidative stress. The current study examined the effects of alterations in the intrarenal dopaminergic system on kidney structure and function in models of type 1 diabetes. We studied catechol-O-methyl-transferase (COMT)(-/-) mice, which have increased renal dopamine production due to decreased dopamine metabolism, and renal transplantation was used to determine whether the effects seen with COMT deficiency were kidney-specific. To determine the effects of selective inhibition of intrarenal dopamine production, we used mice with proximal tubule deletion of aromatic amino acid decarboxylase (ptAADC(-/-)). Compared with wild-type diabetic mice, COMT(-/-) mice had decreased hyperfiltration, decreased macula densa cyclooxygenase-2 expression, decreased albuminuria, decreased glomerulopathy, and inhibition of expression of markers of inflammation, oxidative stress, and fibrosis. These differences were also seen in diabetic mice with a transplanted kidney from COMT(-/-) mice. In contrast, diabetic ptAADC(-/-) mice had increased nephropathy. Our study demonstrates an important role of the intrarenal dopaminergic system to modulate the development and progression of diabetic kidney injury and indicate that the decreased renal dopamine production may have important consequences in the underlying pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
- Ming-Zhi Zhang
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
- Corresponding author: Ming-Zhi Zhang, , or Raymond C. Harris,
| | - Bing Yao
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Shilin Yang
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Suwan Wang
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Xiaofeng Fan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Huiyong Yin
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Agnes B. Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Gilbert W. Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Raymond C. Harris
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
- Nashville Veterans Affairs Hospital, Nashville, Tennessee
- Corresponding author: Ming-Zhi Zhang, , or Raymond C. Harris,
| |
Collapse
|
47
|
Moreira-Rodrigues M, Quelhas-Santos J, Roncon-Albuquerque R, Serrão P, Leite-Moreira A, Sampaio-Maia B, Pestana M. Blunted renal dopaminergic system in a mouse model of diet-induced obesity. Exp Biol Med (Maywood) 2012; 237:949-55. [DOI: 10.1258/ebm.2012.012077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Obesity has reached epidemic proportions in the Western world and is implicated in the pathophysiology of essential hypertension. The aim of the present study was to evaluate sodium handling, blood pressure and renal dopaminergic system activity in a mouse model of obesity induced by exposure to a hypercaloric diet. From six to 18 weeks of age, animals were fed with a control diet or a high-fat high-simple-carbohydrate (HFHSC) diet. Renal function, blood pressure and urinary and plasmatic catecholamines and biochemical parameters were evaluated in both groups. In parallel, the effects of high sodium intake (HS, 1.0% NaCl, 3 days) on natriuresis, urinary catecholamine excretion and aromatic l-amino acid decarboxylase (AADC) activity were evaluated in control and obese mice. Mice exposed to the HFHSC diet presented obesity, hyperglycemia, glucose intolerance, insulin resistance, hyperinsulinemia and increased blood pressure. This was accompanied, in obese mice, by decreases in urinary excretion of dopamine and metabolites as well as reduced AADC activity in renal tissues. During HS intake, absolute urinary dopamine excretion increased in control, but not in obese mice. This was accompanied in obese mice by a natriuretic resistance on day 1 of the HS diet. In addition, obese mice presented increased urinary and plasmatic noradrenaline levels, as well as an increased heart rate when compared with control mice. In conclusion, in this model of diet-induced obesity hyperinsulinemia, insulin resistance and increased sympathetic tone are associated with blunted renal dopaminergic activity. It is suggested that this may contribute to compromised sodium excretion and increased blood pressure in obesity.
Collapse
Affiliation(s)
- Mónica Moreira-Rodrigues
- Nephrology Research and Development Unit, Faculty of Medicine
- Neuropharmacology, IBMC
- Department of Pharmacology and Therapeutics, Faculty of Medicine
| | | | | | - Paula Serrão
- Department of Pharmacology and Therapeutics, Faculty of Medicine
| | | | - Benedita Sampaio-Maia
- Nephrology Research and Development Unit, Faculty of Medicine
- Faculty of Dental Medicine, University of Porto, 4200 Porto
| | - Manuel Pestana
- Nephrology Research and Development Unit, Faculty of Medicine
- Department of Nephrology, Hospital S. João, 4200–319 Porto, Portugal
| |
Collapse
|
48
|
Damkjaer M, Vafaee M, Braad PE, Petersen H, Høilund-Carlsen PF, Bie P. Renal cortical and medullary blood flow during modest saline loading in humans. Acta Physiol (Oxf) 2012; 205:472-83. [PMID: 22433079 DOI: 10.1111/j.1748-1716.2012.02436.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/05/2012] [Accepted: 03/13/2012] [Indexed: 02/02/2023]
Abstract
AIM Renal medullary blood flow (RMBF) is considered an important element of sodium homeostasis, but the experimental evidence is incongruent. Studies in anaesthetized animals generally support the concept in contrast to measurements in conscious animals. We hypothesized that saline-induced natriuresis is associated with changes in RMBF in humans. METHODS After 4 days of low-sodium diet, healthy men were subjected to slow intravenous saline loading (12 μmol kg(-1) min(-1)) for 4 h. Renal medullary and cortical blood flow was determined by positron emission tomography with H(2)(15)O before and after saline infusion using two independent imaging processing methods. One based on a previously published algorithm (voxel peeling) and a novel method based on contrast-enhanced computed tomography (CT). Blood pressure was measured oscillometrically every 10 min. Cardiac output, heart rate and total peripheral resistance were recorded continuously. RESULTS Saline loading increased the urinary sodium excretion by 3.6-fold (21-76 μmol min(-1) , P < 0.01). The RMBF was 2.6 ± 0.2 mL g(-1) tissue min(-1) before and 2.7 ± 0.1 mL g(-1) tissue min(-1) after saline (n.s.). Cortical blood flow was 3.6 ± 0.1 before and 3.4 ± 0.2 after saline (n.s.). Mean arterial blood pressure did not change measurably (90 vs. 90 mmHg). Bland-Altman analysis suggested agreement between results obtained with voxel peeling (2.6 ± 0.2 mL g(-1) tissue min(-1)) and contrast-enhanced CT (2.0 ± 0.1 mL g(-1) tissue min(-1)). CONCLUSION In normal humans, changes in RMBF are not necessarily involved in the natriuretic response to modest saline loading. This result is in line with data from conscious rodents.
Collapse
Affiliation(s)
- M. Damkjaer
- Institute of Molecular Medicine; University of Southern Denmark; Odense; Denmark
| | - M. Vafaee
- Department of Neuroscience and Pharmacology; Faculty of Health Sciences; University of Copenhagen; Copenhagen; Denmark
| | - P. E. Braad
- Department of Nuclear Medicine; Odense University Hospital; Odense; Denmark
| | - H. Petersen
- Department of Nuclear Medicine; Odense University Hospital; Odense; Denmark
| | | | - P. Bie
- Institute of Molecular Medicine; University of Southern Denmark; Odense; Denmark
| |
Collapse
|
49
|
Moura E, Silva E, Serrão MP, Afonso J, Kozmus CEP, Vieira-Coelho MA. α2C-Adrenoceptors modulate L-DOPA uptake in opossum kidney cells and in the mouse kidney. Am J Physiol Renal Physiol 2012; 303:F928-38. [PMID: 22859407 DOI: 10.1152/ajprenal.00217.2011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Targeted deletion or selective pharmacological inhibition of α(2C)-adrenoceptors in mice results in increased brain tissue levels of dopamine and its precursor l-3,4-dihydroxyphenylalanine (l-DOPA), without significant changes in l-DOPA synthesis. l-DOPA uptake is considered the rate-limiting step in dopamine synthesis in the kidney. Since α(2C)-adrenoceptors may influence the transport of l-DOPA, we investigated the effect of α(2C)-adrenoceptor activation on l-DOPA uptake in a kidney cell line (opossum kidney cells). l-DOPA and dopamine kidney tissue levels in α(2C)-adrenoceptor knockout (α(2C)KO) mice and in mice treated with the selective α(2C)-adrenoceptor antagonist JP-1302 were also evaluated. The α(2)-adrenoceptor agonist medetomidine (0.1-1,000 nM) produced a concentration-dependent decrease in l-DOPA uptake in opossum kidney cells (IC(50): 2.5 ± 0.5 nM and maximal effect: 28 ± 5% of inhibition). This effect was abolished by a preincubation with JP-1302 (300 nM). Furthermore, the effect of medetomidine (100 nM) was abolished by a preincubation with U-0126 (10 μM), a MEK1/2 inhibitor. Kidney tissue levels of l-DOPA were significantly higher in α(2C)KO mice compared with wild-type mice (wild-type mice: 58 ± 2 pmol/g tissue and α(2C)KO mice: 81 ± 15 pmol/g tissue, P < 0.05) and in mice treated with JP-1302 (3 μmol/kg body wt) compared with control mice (control mice: 62 ± 2 pmol/g tissue and JP-1302-treated mice: 75 ± 1 pmol/g tissue, P < 0.05), both without significant changes in dopamine kidney tissue levels. However, mice treated with JP-1302 on a high-salt diet presented significantly higher dopamine levels in the kidney and urine compared with control animals on a high-salt diet. In conclusion, in a kidney cell line, α(2C)-adrenoceptor activation inhibits l-DOPA uptake, and in mice, deletion or blockade of α(2C)-adrenoceptors increases l-DOPA kidney tissue levels.
Collapse
Affiliation(s)
- Eduardo Moura
- Departamento de Farmacologia e Terapêutica, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, Porto 4200-319, Portugal.
| | | | | | | | | | | |
Collapse
|
50
|
Yang Y, Zhang Y, Cuevas S, Villar VA, Escano C, Asico L, Yu P, Grandy DK, Felder RA, Armando I, Jose PA. Paraoxonase 2 decreases renal reactive oxygen species production, lowers blood pressure, and mediates dopamine D2 receptor-induced inhibition of NADPH oxidase. Free Radic Biol Med 2012; 53:437-46. [PMID: 22634053 PMCID: PMC3408834 DOI: 10.1016/j.freeradbiomed.2012.05.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 04/20/2012] [Accepted: 05/09/2012] [Indexed: 12/22/2022]
Abstract
The dopamine D(2) receptor (D(2)R) regulates renal reactive oxygen species (ROS) production, and impaired D(2)R function results in ROS-dependent hypertension. Paraoxonase 2 (PON2), which belongs to the paraoxonase gene family, is expressed in various tissues, acting to protect against cellular oxidative stress. We hypothesized that PON2 may be involved in preventing excessive renal ROS production and thus may contribute to maintenance of normal blood pressure. Moreover, D(2)R may decrease ROS production, in part, through regulation of PON2. D(2)R colocalized with PON2 in the brush border of mouse renal proximal tubules. Renal PON2 protein was decreased (-33±6%) in D(2)(-/-) relative to D(2)(+/+) mice. Renal subcapsular infusion of PON2 siRNA decreased PON2 protein expression (-55%), increased renal oxidative stress (2.2-fold), associated with increased renal NADPH oxidase expression (Nox1, 1.9-fold; Nox2, 2.9-fold; and Nox4, 1.6-fold) and activity (1.9-fold), and elevated arterial blood pressure (systolic, 134±5 vs 93±6mmHg; diastolic, 97±4 vs 65±7mmHg; mean 113±4 vs 75±7mmHg). To determine the relevance of the PON2 and D(2)R interaction in humans, we studied human renal proximal tubule cells. Both D(2)R and PON2 were found in nonlipid and lipid rafts and physically interacted with each other. Treatment of these cells with the D(2)R/D(3)R agonist quinpirole (1μM, 24h) decreased ROS production (-35±6%), associated with decreased NADPH oxidase activity (-32±3%) and expression of Nox2 (-41±7%) and Nox4 (-47±8%) protein, and increased expression of PON2 mRNA (2.1-fold) and protein (1.6-fold) at 24h. Silencing PON2 (siRNA, 10nM, 48h) not only partially prevented the quinpirole-induced decrease in ROS production by 36%, but also increased basal ROS production (1.3-fold), which was associated with an increase in NADPH oxidase activity (1.4-fold) and expression of Nox2 (2.1-fold) and Nox4 (1.8-fold) protein. Inhibition of NADPH oxidase with diphenylene iodonium (10μM/30 min) inhibited the increase in ROS production caused by PON2 silencing. Our results suggest that renal PON2 is involved in the inhibition of renal NADPH oxidase activity and ROS production and contributes to the maintenance of normal blood pressure. PON2 is positively regulated by D(2)R and may, in part, mediate the inhibitory effect of renal D(2)R on NADPH oxidase activity and ROS production.
Collapse
Affiliation(s)
- Yu Yang
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| | - Yanrong Zhang
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| | - Santiago Cuevas
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| | - Van Anthony Villar
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| | - Crisanto Escano
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| | - Laureano Asico
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| | - Peiying Yu
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| | - David K. Grandy
- Departments of Physiology and Pharmacology, Oregon Health and Sciences University, Portland, OR 97239
| | - Robin A. Felder
- Department of Pathology, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - Ines Armando
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
- Corresponding author. Fax: 202-476-6582, (I.Armando)
| | - Pedro A. Jose
- Center for Molecular Physiology Research, Children's National Medical Center, George Washington University, Washington, DC 20010
| |
Collapse
|