1
|
You R, Jia Z. Pathophysiological role of Na-Cl cotransporter in kidneys, blood pressure, and metabolism. Hum Cell 2024; 37:1306-1315. [PMID: 38985392 DOI: 10.1007/s13577-024-01099-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
The Na-Cl cotransporter (NCC) is a well-recognized regulator of ion transportation in the kidneys that facilitates Na+ reabsorption in the distal convoluted tubule. It is also the pharmacologic inhibitory target of thiazide diuretics, a class of front-line antihypertensive agents that have been widely used for decades. NCC is a potent regulator of Na+ reabsorption and homeostasis. Hence, its overactivation and suppression lead to hypertension and hypotension, respectively. Genetic mutations that affect NCC function contribute to several diseases such as Gordon and Gitelman syndromes. We summarized the role of NCC in various physiologic processes and pathological conditions, such as maintaining ion and water homeostasis, controlling blood pressure, and influencing renal physiology and injury. In addition, we discussed the recent advancements in understanding cryo-EM structure of NCC, the regulatory mechanisms and binding mode of thiazides with NCC, and novel physiologic implications of NCC in regulating the cross-talk between the immune system and adipose tissue or the kidneys. This review contributes to a comprehensive understanding of the pivotal role of NCC in maintaining ion homeostasis, regulating blood pressure, and facilitating kidney function and NCC's novel role in immune and metabolic regulation.
Collapse
Affiliation(s)
- Ran You
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, China.
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Li H, Wan L, Liu M, Ma E, Huang L, Yang Y, Li Q, Fang Y, Li J, Han B, Zhang C, Sun L, Hou X, Li H, Sun M, Qian S, Duan X, Zhao R, Yang X, Chen Y, Wu S, Zhang X, Zhang Y, Cheng G, Chen G, Gao Q, Xu J, Hou L, Wei C, Zhong H. SARS-CoV-2 spike-induced syncytia are senescent and contribute to exacerbated heart failure. PLoS Pathog 2024; 20:e1012291. [PMID: 39102426 PMCID: PMC11326701 DOI: 10.1371/journal.ppat.1012291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 08/15/2024] [Accepted: 05/27/2024] [Indexed: 08/07/2024] Open
Abstract
SARS-CoV-2 spike protein (SARS-2-S) induced cell-cell fusion in uninfected cells may occur in long COVID-19 syndrome, as circulating SARS-2-S or extracellular vesicles containing SARS-2-S (S-EVs) were found to be prevalent in post-acute sequelae of COVID-19 (PASC) for up to 12 months after diagnosis. Although isolated recombinant SARS-2-S protein has been shown to increase the SASP in senescent ACE2-expressing cells, the direct linkage of SARS-2-S syncytia with senescence in the absence of virus infection and the degree to which SARS-2-S syncytia affect pathology in the setting of cardiac dysfunction are unknown. Here, we found that the senescent outcome of SARS-2-S induced syncytia exacerbated heart failure progression. We first demonstrated that syncytium formation in cells expressing SARS-2-S delivered by DNA plasmid or LNP-mRNA exhibits a senescence-like phenotype. Extracellular vesicles containing SARS-2-S (S-EVs) also confer a potent ability to form senescent syncytia without de novo synthesis of SARS-2-S. However, it is important to note that currently approved COVID-19 mRNA vaccines do not induce syncytium formation or cellular senescence. Mechanistically, SARS-2-S syncytia provoke the formation of functional MAVS aggregates, which regulate the senescence fate of SARS-2-S syncytia by TNFα. We further demonstrate that senescent SARS-2-S syncytia exhibit shrinked morphology, leading to the activation of WNK1 and impaired cardiac metabolism. In pre-existing heart failure mice, the WNK1 inhibitor WNK463, anti-syncytial drug niclosamide, and senolytic dasatinib protect the heart from exacerbated heart failure triggered by SARS-2-S. Our findings thus suggest a potential mechanism for COVID-19-mediated cardiac pathology and recommend the application of WNK1 inhibitor for therapy especially in individuals with post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Huilong Li
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luming Wan
- Beijing Institute of Biotechnology, Beijing, China
| | - Muyi Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Enhao Ma
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Linfei Huang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yilong Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Qihong Li
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yi Fang
- The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jingfei Li
- Beijing Institute of Biotechnology, Beijing, China
| | - Bingqing Han
- Beijing Institute of Biotechnology, Beijing, China
| | - Chang Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Lijuan Sun
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Xufeng Hou
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Haiyang Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Mingyu Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Sichong Qian
- Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xuejing Duan
- Department of Pathology, Fuwai Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing, China
| | - Ruzhou Zhao
- Beijing Institute of Biotechnology, Beijing, China
| | - Xiaopan Yang
- Beijing Institute of Biotechnology, Beijing, China
| | - Yi Chen
- Beijing Institute of Biotechnology, Beijing, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Xuhui Zhang
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | | | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Gengye Chen
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qi Gao
- Beijing Yaogen Biotechnology Co.Ltd, Beijing, China
| | - Junjie Xu
- Beijing Institute of Biotechnology, Beijing, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, Beijing, China
- College of Basic Medical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Congwen Wei
- Beijing Institute of Biotechnology, Beijing, China
| | - Hui Zhong
- Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
3
|
Rioux AV, Nsimba-Batomene TR, Slimani S, Bergeron NAD, Gravel MAM, Schreiber SV, Fiola MJ, Haydock L, Garneau AP, Isenring P. Navigating the multifaceted intricacies of the Na +-Cl - cotransporter, a highly regulated key effector in the control of hydromineral homeostasis. Physiol Rev 2024; 104:1147-1204. [PMID: 38329422 PMCID: PMC11381001 DOI: 10.1152/physrev.00027.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/01/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
The Na+-Cl- cotransporter (NCC; SLC12A3) is a highly regulated integral membrane protein that is known to exist as three splice variants in primates. Its primary role in the kidney is to mediate the cosymport of Na+ and Cl- across the apical membrane of the distal convoluted tubule. Through this role and the involvement of other ion transport systems, NCC allows the systemic circulation to reclaim a fraction of the ultrafiltered Na+, K+, Cl-, and Mg+ loads in exchange for Ca2+ and [Formula: see text]. The physiological relevance of the Na+-Cl- cotransport mechanism in humans is illustrated by several abnormalities that result from NCC inactivation through the administration of thiazides or in the setting of hereditary disorders. The purpose of the present review is to discuss the molecular mechanisms and overall roles of Na+-Cl- cotransport as the main topics of interest. On reading the narrative proposed, one will realize that the knowledge gained in regard to these themes will continue to progress unrelentingly no matter how refined it has now become.
Collapse
Affiliation(s)
- A V Rioux
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - T R Nsimba-Batomene
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S Slimani
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - N A D Bergeron
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M A M Gravel
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S V Schreiber
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M J Fiola
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - L Haydock
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - A P Garneau
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - P Isenring
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
4
|
Abstract
Salt (sodium chloride) is an essential nutrient required to maintain physiological functions. However, for most people, daily salt intake far exceeds their physiological need and is habitually greater than recommended upper thresholds. Excess salt intake leads to elevation in blood pressure which drives cardiovascular morbidity and mortality. Indeed, excessive salt intake is estimated to be responsible for ≈5 million deaths per year globally. For approximately one-third of otherwise healthy individuals (and >50% of those with hypertension), the effect of salt intake on blood pressure elevation is exaggerated; such people are categorized as salt sensitive and salt sensitivity of blood pressure is considered an independent risk factor for cardiovascular disease and death. The prevalence of salt sensitivity is higher in women than in men and, in both, increases with age. This narrative review considers the foundational concepts of salt sensitivity and the underlying effector systems that cause salt sensitivity. We also consider recent updates in preclinical and clinical research that are revealing new modifying factors that determine the blood pressure response to high salt intake.
Collapse
Affiliation(s)
- Matthew A Bailey
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom (M.A.B., N.D.)
| | - Neeraj Dhaun
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom (M.A.B., N.D.)
- Department of Renal Medicine, Royal Infirmary of Edinburgh, United Kingdom (N.D.)
| |
Collapse
|
5
|
Wen Y, Lu X, Privratsky JR, Ren J, Ali S, Yang B, Rudemiller NP, Zhang J, Nedospasov SA, Crowley SD. TNF- α from the Proximal Nephron Exacerbates Aristolochic Acid Nephropathy. KIDNEY360 2024; 5:44-56. [PMID: 37986166 PMCID: PMC10833606 DOI: 10.34067/kid.0000000000000314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Key Points Proximal tubular TNF aggravates kidney injury and fibrogenesis in aristolochic acid nephropathy. Tubular TNF disrupts the cell cycle in injured tubular epithelial cells. TNF-mediated toxic renal injury is independent of systemic immune responses. Background Aristolochic acid nephropathy (AAN) presents with tubular epithelial cell (TEC) damage and tubulointerstitial inflammation. Although TNF-α regulates cell apoptosis and inflammatory responses, the effects of tubular TNF in the progression of AAN require elucidation. Methods Floxed TNF mice on the 129/SvEv background were crossed with PEPCK-Cre mice to generate PEPCK-Cre + TNF flox/flox (TNF PTKO) mice or bred with Ksp-Cre mice to generate KSP-Cre + TNF flox/flox (TNF DNKO) mice. TNF PTKO, TNF DNKO, and wild-type controls (Cre negative littermates) were subjected to acute and chronic AAN. Results Deletion of TNF in the proximal but not distal nephron attenuated kidney injury, renal inflammation, and tubulointerstitial fibrosis after acute or chronic aristolochic acid (AA) exposure. The TNF PTKO mice did not have altered numbers of infiltrating myeloid cells in AAN kidneys. Nevertheless, kidneys from AA-treated TNF PTKO mice had reduced levels of proteins involved in regulated cell death, higher proportions of TECs in the G0/G1 phase, and reduced TEC proportions in the G2/M phase. Pifithrin-α , which restores the cell cycle, abrogated differences between the wild-type and PTKO cohorts in G2/M phase arrest of TECs and kidney fibrosis after AA exposure. Conclusions TNF from the proximal but not the distal nephron propagates kidney injury and fibrogenesis in AAN in part by inducing G2/M cell cycle arrest of TECs.
Collapse
Affiliation(s)
- Yi Wen
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
- Department of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaohan Lu
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Jamie R. Privratsky
- Department of Anesthesiology, Durham VA and Duke University Medical Center, Durham, North Carolina
| | - Jiafa Ren
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Saba Ali
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Bo Yang
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Nathan P. Rudemiller
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| | - Jiandong Zhang
- Division of Cardiology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sergei A. Nedospasov
- Engelhardt Institute of Molecular Biology, Moscow, Russia
- Institute of Cell Biology and Neurobiology, Universitatsmedizin, Berlin, Germany
| | - Steven D. Crowley
- Division of Nephrology, Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina
| |
Collapse
|
6
|
Valga F, Monzon T, Vega-Diaz N, Santana A, Moscol G, Ruiz-Santana S, Rodriguez-Perez JC. Serum chloride as a marker of cardiovascular and all-cause mortality in chronic hemodialysis patients: 5-Year follow-up study. Nefrologia 2023; 43 Suppl 2:47-56. [PMID: 38245440 DOI: 10.1016/j.nefroe.2023.05.009] [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: 08/23/2022] [Accepted: 02/20/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Hypochloremia has been associated with increased mortality in patients with hypertension, heart failure, sepsis, and chronic kidney disease (CKD). The pathophysiological mechanisms of this finding are not clear. There are no studies describing an association between serum chloride levels (Cl-) and mortality in incident chronic hemodialysis (HD) patients. METHOD Retrospective cohort study of the incident population in our chronic outpatient hemodialysis program between January 1, 2016, and January 1, 2021 (N=374). Survival time was collected in all patients and analyzed using the Kaplan-Meyer method. A multivariate Cox regression model was performed to predict the probability of survival, applying a stepwise procedure. RESULTS During the median follow-up period of 20 months, 83 patients died. The 5-year overall survival rate for our patients was 45%. Both natremia and chloremia had no significant differences when compared by sex, vascular access, or etiology. There was an inverse correlation between Cl- and interdialytic weight gain (r=-0.15) (p=0.0038). Patients belonging to the quartile with lower Cl- levels had less probability of survival than patients in the quartile with higher Cl- levels (27% and 68%, respectively, p=0.019). On the other hand, in the multivariate Cox regression model, variables significantly associated with higher mortality were being older, having higher baseline comorbidity by modified Charlson index, not taking diuretics and having lower albumin and chloride levels. Particularly, higher Cl- levels was independently associated with both lower all-cause mortality (adjusted hazard ratio [HR]=0.84; 95% confidence interval [CI], 0.77-0.92; p=0.0001) and cardiovascular mortality (HR 0.9; 95% CI, 0.83-0.97; p<0.0057). CONCLUSIONS Lower Cl- levels were associated with higher all-cause and cardiovascular mortality in incident patients on chronic hemodialysis in our health area.
Collapse
Affiliation(s)
- Francisco Valga
- Nephrology Department, Doctor Negrin University Hospital of Gran Canaria, Las Palmas de Gran Canaria, Spain; Doctoral School, Biomedicine Research Program, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.
| | - Tania Monzon
- Nephrology Department, Doctor Negrin University Hospital of Gran Canaria, Las Palmas de Gran Canaria, Spain; Doctoral School, Biomedicine Research Program, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Nicanor Vega-Diaz
- Doctoral School, Biomedicine Research Program, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain; Avericum S.L Hemodialysis Centers, Las Palmas de Gran Canaria, Spain
| | - Angelo Santana
- Mathematics Department, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Giancarlo Moscol
- Oncology Department, MD Anderson Cancer Center, Houston, TX, USA
| | - Sergio Ruiz-Santana
- Doctoral School, Biomedicine Research Program, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain; Intensive Care Unit, Doctor Negrin University Hospital of Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Jose Carlos Rodriguez-Perez
- Nephrology Department, Doctor Negrin University Hospital of Gran Canaria, Las Palmas de Gran Canaria, Spain; University Fernando Pessoa-Canarias, Las Palmas, Spain
| |
Collapse
|
7
|
Matsuki H, Mandai S, Shiwaku H, Koide T, Takahashi N, Yanagi T, Inaba S, Ida S, Fujiki T, Mori Y, Ando F, Mori T, Susa K, Iimori S, Sohara E, Takahashi H, Uchida S. Chronic kidney disease causes blood-brain barrier breakdown via urea-activated matrix metalloproteinase-2 and insolubility of tau protein. Aging (Albany NY) 2023; 15:10972-10995. [PMID: 37889501 PMCID: PMC10637825 DOI: 10.18632/aging.205164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Chronic kidney disease (CKD) causes cognitive impairment and contributes to the overall global burden of dementia. However, mechanisms through which the kidneys and brain communicate are not fully understood. We established a CKD mouse model through adenine-induced tubulointerstitial fibrosis. Novel object recognition tests indicated that CKD decreased recognition memory. Sarkosyl-insoluble-proteomic analyses of the CKD mouse hippocampus revealed an accumulation of insoluble MAPT (microtubule-associated protein tau) and RNA-binding proteins such as small nuclear ribonucleoprotein U1 subunit 70 (SNRNP70). Additionally, there was an accumulation of Immunoglobulin G (IgG), indicating blood-brain barrier (BBB) breakdown. We identified that expressions of essential tight-junction protein claudin-5 and adherens-junction protein platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31) were decreased in the brain endothelial cells of CKD mice. We determined urea as a major uremic solute that dose dependently decreased both claudin-5 and PECAM-1 expression in the mouse brain endothelial cell line bEnd.3 cells. Gelatin zymography indicated that the serum of CKD mice activated matrix metalloproteinase-2 (MMP2), while marimastat ameliorated the reduction of claudin-5 expression by urea in bEnd.3 cells. This study established a brain proteomic signature of CKD indicating BBB breakdown and insolubility of tau protein, which are pathologically linked to Alzheimer's disease. Urea-mediated activation of MMP2 was partly responsible for BBB breakdown in CKD.
Collapse
Affiliation(s)
- Hisazumi Matsuki
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Shintaro Mandai
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Hiroki Shiwaku
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Takaaki Koide
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Naohiro Takahashi
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Tomoki Yanagi
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Shunsuke Inaba
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Saaya Ida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Tamami Fujiki
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Yutaro Mori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Fumiaki Ando
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Takayasu Mori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Koichiro Susa
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Soichiro Iimori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Hidehiko Takahashi
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
- Center for Brain Integration Research, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo City, Tokyo 113-8519, Japan
| |
Collapse
|
8
|
Akhouri V, Majumder S, Gaikwad AB. The emerging insight into E3 ligases as the potential therapeutic target for diabetic kidney disease. Life Sci 2023; 321:121643. [PMID: 36997061 DOI: 10.1016/j.lfs.2023.121643] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/25/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023]
Abstract
Diabetic kidney disease (DKD) is a major diabetic complication and global health concern, occurring in nearly 30 % to 40 % of people with diabetes. Importantly, several therapeutic strategies are being used against DKD; however, available treatments are not uniformly effective and the continuous rise in the prevalence of DKD demands more potential therapeutic approaches or targets. Epigenetic modifiers are regarded for their potential therapeutic effects against DKD. E3 ligases are such epigenetic modifier that regulates the target gene expression by attaching ubiquitin to the histone protein. In recent years, the E3 ligases came up as a potential therapeutic target as it selectively attaches ubiquitin to the substrate proteins in the ubiquitination cascade and modulates cellular homeostasis. The E3 ligases are also actively involved in DKD by regulating the expression of several proteins involved in the proinflammatory and profibrotic pathways. Burgeoning reports suggest that several E3 ligases such as TRIM18 (tripartite motif 18), Smurf1 (Smad ubiquitination regulatory factor 1), and NEDD4-2 (neural precursor cell-expressed developmentally downregulated gene 4-2) are involved in kidney epithelial-mesenchymal transition, inflammation, and fibrosis by regulating respective signaling pathways. However, the various signaling pathways that are regulated by different E3 ligases in the progression of DKD are poorly understood. In this review, we have discussed E3 ligases as potential therapeutic target for DKD. Moreover, different signaling pathways regulated by E3 ligases in the progression of DKD have also been discussed.
Collapse
Affiliation(s)
- Vivek Akhouri
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Syamantak Majumder
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
9
|
Uchida S, Mori T, Susa K, Sohara E. NCC regulation by WNK signal cascade. Front Physiol 2023; 13:1081261. [PMID: 36685207 PMCID: PMC9845728 DOI: 10.3389/fphys.2022.1081261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023] Open
Abstract
With-no-lysine (K) (WNK) kinases have been identified as the causal genes for pseudohypoaldosteronism type II (PHAII), a rare hereditary hypertension condition characterized by hyperkalemia, hyperchloremic metabolic acidosis, and thiazide-hypersensitivity. We thought that clarifying the link between WNK and NaCl cotransporter (NCC) would bring us new mechanism(s) of NCC regulation. For the first time, we were able to produce a knock-in mouse model of PHAII and anti-phosphorylated NCC antibodies against the putative NCC phosphorylation sites and discover that constitutive activation of NCC and increased phosphorylation of NCC are the primary pathogenesis of the disease in vivo. We have since demonstrated that this regulatory mechanism is mediated by the kinases oxidative stress-response protein 1 (OSR1) and STE20/SPS1-related proline/alanine-rich kinase (SPAK) (WNK-OSR1/SPAK-NCC signaling cascade) and that the signaling is not only important in the pathological condition of PHAII but also plays a crucial physiological role in the regulation of NCC.
Collapse
|
10
|
Peng Z, Xu Q, Hu W, Cheng Y. Review on Molecular Mechanism of Hypertensive Nephropathy. Curr Pharm Des 2023; 29:2568-2578. [PMID: 37927071 DOI: 10.2174/0113816128266582231031111516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023]
Abstract
Hypertension, a prevalent chronic ailment, has the potential to impair kidney function, and thereby resulting in hypertensive nephropathy. The escalating incidence of hypertensive nephropathy attributed to the aging population in urban areas, has emerged as a prominent cause of end-stage renal disease. Nevertheless, the intricate pathogenesis of hypertensive nephropathy poses considerable obstacles in terms of precise clinical diagnosis and treatment. This paper aims to consolidate the research findings on the pathogenesis of hypertensive nephropathy by focusing on the perspective of molecular biology.
Collapse
Affiliation(s)
- Zhen Peng
- Department of Pharmacy, Yichun People's Hospital, Yichun, Jiangxi 336000, China
| | - Qiaohong Xu
- Department of Pharmacy, Yichun People's Hospital, Yichun, Jiangxi 336000, China
| | - Wen Hu
- Department of Pharmacy, Yichun People's Hospital, Yichun, Jiangxi 336000, China
| | - Yimin Cheng
- Jiangxi Provincial Key Laboratory of Natural Active Pharmaceutical Constituents, Department of Chemistry and Bioengineering, Yichun University, Yichun, Jiangxi 336000, China
| |
Collapse
|
11
|
Molecular Mechanisms of Na-Cl Cotransporter in Relation to Hypertension in Chronic Kidney Disease. Int J Mol Sci 2022; 24:ijms24010286. [PMID: 36613730 PMCID: PMC9820686 DOI: 10.3390/ijms24010286] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic kidney disease (CKD) is a common clinical disease with an increasing incidence, affecting 10 to 15% of the world's population. Hypertension is the most common and modifiable risk factor for preventing adverse cardiovascular outcomes in patients with CKD. A survey from developed countries shows that 47% of hypertensive patients over the age of 20 have uncontrolled blood pressure (BP), and the control rate is even lower in developing countries. CKD is both a common cause of uncontrolled hypertension and a risk factor for altered sequelae. In particular, studies have demonstrated that abnormal blood-pressure patterns in CKD patients, such as non-dipping-blood-pressure patterns, are associated with a significantly increased risk of cardiovascular (CV) disease. The distal convoluted tubule (DCT) is a region of the kidney, and although only 5-10% of the sodium (Na+) filtered by the glomerulus is reabsorbed by DCT, most studies agree that Na-Cl cotransporter (NCC) in human, rabbit, mouse, and rat kidneys is the most important route of sodium reabsorption across the DCT for maintaining the homeostasis of sodium. The regulation of NCC involves a large and complex network structure, including certain physiological factors, kinases, scaffold proteins, transporter phosphorylation, and other aspects. This regulation network includes various levels. Naturally, cross-talk between the components of this system must occur in order to relay the important signals to the transporter to play its role. Knowledge of the mechanisms regulating NCC activation is critical for understanding and treating hypertension and CKD. Previous studies from our laboratory have investigated the mechanisms through which NCC is activated in several different models. In the following sections, we review the literature on the mechanisms of NCC in relation to hypertension in CKD.
Collapse
|
12
|
Ishii T, Mimura I, Nagaoka K, Naito A, Sugasawa T, Kuroda R, Yamada D, Kanki Y, Kume H, Ushiku T, Kakimi K, Tanaka T, Nangaku M. Effect of M2-like macrophages of the injured-kidney cortex on kidney cancer progression. Cell Death Dis 2022; 8:480. [PMID: 36470862 PMCID: PMC9722672 DOI: 10.1038/s41420-022-01255-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) affects kidney cancer patients' mortality. However, the underlying mechanism remains unknown. M2-like macrophages have pro-tumor functions, also exist in injured kidney, and promote kidney fibrosis. Thus, it is suspected that M2-like macrophages in injured kidney induce the pro-tumor microenvironment leading to kidney cancer progression. We found that M2-like macrophages present in the injured kidney promoted kidney cancer progression and induced resistance to anti-PD1 antibody through its pro-tumor function and inhibition of CD8+ T cell infiltration. RNA-seq revealed Slc7a11 was upregulated in M2-like macrophages. Inhibition of Slc7a11 with sulfasalazine inhibited the pro-tumor function of M2-like macrophages and synergized with anti-PD1 antibody. Moreover, SLC7A11-positive macrophages were associated with poor prognosis among kidney cancer patients. Collectively, this study dissects the characteristic microenvironment in the injured kidney that contributed to kidney cancer progression and anti-PD1 antibody resistance. This insight offers promising combination therapy with anti-PD1 antibody and macrophage targeted therapy.
Collapse
Affiliation(s)
- Taisuke Ishii
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Imari Mimura
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Koji Nagaoka
- grid.412708.80000 0004 1764 7572Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Akihiro Naito
- grid.26999.3d0000 0001 2151 536XDivision of Urology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Takehito Sugasawa
- grid.20515.330000 0001 2369 4728Laboratory of Clinical Examination/Sports Medicine, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 3058577 Japan
| | - Ryohei Kuroda
- grid.26999.3d0000 0001 2151 536XDepartment of Pathology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Daisuke Yamada
- grid.26999.3d0000 0001 2151 536XDivision of Urology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Yasuharu Kanki
- grid.20515.330000 0001 2369 4728Laboratory of Clinical Examination/Sports Medicine, Division of Clinical Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 3058577 Japan
| | - Haruki Kume
- grid.26999.3d0000 0001 2151 536XDivision of Urology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Tetsuo Ushiku
- grid.26999.3d0000 0001 2151 536XDepartment of Pathology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Kazuhiro Kakimi
- grid.412708.80000 0004 1764 7572Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| | - Tetsuhiro Tanaka
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan ,grid.69566.3a0000 0001 2248 6943Department of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 9808574 Japan
| | - Masaomi Nangaku
- grid.26999.3d0000 0001 2151 536XDivision of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 1138655 Japan
| |
Collapse
|
13
|
Crorkin P, Hao S, Ferreri NR. Responses to Ang II (Angiotensin II), Salt Intake, and Lipopolysaccharide Reveal the Diverse Actions of TNF-α (Tumor Necrosis Factor-α) on Blood Pressure and Renal Function. Hypertension 2022; 79:2656-2670. [PMID: 36129177 PMCID: PMC9649876 DOI: 10.1161/hypertensionaha.122.19464] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
TNF-α (tumor necrosis factor-alpha) is the best known as a proinflammatory cytokine; yet, this cytokine also has important immunomodulatory and regulatory functions. As the effects of TNF-α on immune system function were being revealed, the spectrum of its activities appeared in conflict with each other before investigators defined the settings and mechanisms by which TNF-α contributed to both host defense and chronic inflammation. These effects reflect self-protective mechanisms that may become harmful when dysregulated. The paradigm of physiological and pathophysiological effects of TNF-α has since been uncovered in the lung, colon, and kidney where its role has been identified in pulmonary edema, electrolyte reabsorption, and blood pressure regulation, respectively. Recent studies on the prohypertensive and inflammatory effects of TNF-α in the cardiovascular system juxtaposed to those related to NaCl and blood pressure homeostasis, the response of the kidney to lipopolysaccharide, and protection against bacterial infections are helping define the mechanisms by which TNF-α modulates distinct functions within the kidney. This review discusses how production of TNF-α by renal epithelial cells may contribute to regulatory mechanisms that not only govern electrolyte excretion and blood pressure homeostasis but also maintain the appropriate local hypersalinity environment needed for optimizing the innate immune response to bacterial infections in the kidney. It is possible that the wide range of effects mediated by TNF-α may be related to severity of disease, amount of inflammation and TNF-α levels, and the specific cell types that produce this cytokine, areas that remain to be investigated further.
Collapse
Affiliation(s)
- Patrick Crorkin
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | - Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | | |
Collapse
|
14
|
Rosenbaek LL, Petrillo F, van Bemmelen MX, Staub O, Murali SK, Fenton RA. The E3 ubiquitin-protein ligase Nedd4-2 regulates the sodium chloride cotransporter NCC but is not required for a potassium-induced reduction of NCC expression. Front Physiol 2022; 13:971251. [PMID: 36160843 PMCID: PMC9490057 DOI: 10.3389/fphys.2022.971251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Na+ and K+ balance is influenced by the activity of the sodium chloride cotransporter NCC in the distal convoluted tubule. NCC activity and abundance are reduced by high extracellular K+. The E3 ubiquitin ligase neural precursor cell expressed developmentally downregulated 4–2 (Nedd4-2) has been proposed as a modulator of NCC abundance. Here, we examined the functional role of Nedd4-2 on NCC regulation and whether Nedd4-2 is important for the effects of high extracellular K+ on NCC. Total and plasma membrane levels of ubiquitylated NCC were lower in NCC-expressing MDCKI cells after Nedd4-2 deletion. NCC and phosphorylated NCC (pT58-NCC) levels were higher after Nedd4-2 deletion, and NCC levels on the plasma membrane were elevated. No significant changes were seen after Nedd4-2 knockdown in the levels of SPAK and phosphorylated SPAK (pS373-SPAK), the major NCC regulatory kinase. Nedd4-2 deficiency had no effect on the internalization rate of NCC from the plasma membrane, but NCC protein half-life was increased. In ex vivo experiments with kidney tubule suspensions from Nedd4-2 knockout (KO) mice, high K+ reduced total and pT58-NCC regardless of genotype. We conclude that Nedd4-2 is involved in ubiquitylation of NCC and modulating its plasma membrane levels and degradation. However, Nedd4-2 does not appear to be important for K+ induced reductions in NCC abundance.
Collapse
Affiliation(s)
- Lena L. Rosenbaek
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Leducq Foundation Potassium in Hypertension International Network, Massachusetts, United States
| | | | - Miguel X. van Bemmelen
- Leducq Foundation Potassium in Hypertension International Network, Massachusetts, United States
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Olivier Staub
- Leducq Foundation Potassium in Hypertension International Network, Massachusetts, United States
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Sathish K. Murali
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Leducq Foundation Potassium in Hypertension International Network, Massachusetts, United States
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Robert A. Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Leducq Foundation Potassium in Hypertension International Network, Massachusetts, United States
- *Correspondence: Robert A. Fenton,
| |
Collapse
|
15
|
Bhuiyan MIH, Young CB, Jahan I, Hasan MN, Fischer S, Meor Azlan NF, Liu M, Chattopadhyay A, Huang H, Kahle KT, Zhang J, Poloyac SM, Molyneaux BJ, Straub AC, Deng X, Gomez D, Sun D. NF-κB Signaling-Mediated Activation of WNK-SPAK-NKCC1 Cascade in Worsened Stroke Outcomes of Ang II-Hypertensive Mice. Stroke 2022; 53:1720-1734. [PMID: 35272484 PMCID: PMC9038703 DOI: 10.1161/strokeaha.121.038351] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 01/31/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Worsened stroke outcomes with hypertension comorbidity are insensitive to blood pressure-lowering therapies. In an experimental stroke model with comorbid hypertension, we investigated causal roles of ang II (angiotensin II)-mediated stimulation of the brain WNK (with no lysine [K] kinases)-SPAK (STE20/SPS1-related proline/alanine-rich kinase)-NKCC1 (Na-K-Cl cotransporter) complex in worsened outcomes. METHODS Saline- or ang II-infused C57BL/6J male mice underwent stroke induced by permanent occlusion of the distal branches of the middle cerebral artery. Mice were randomly assigned to receive either vehicle dimethyl sulfoxide/PBS (2 mL/kg body weight/day, IP), a novel SPAK inhibitor, 5-chloro-N-(5-chloro-4-((4-chlorophenyl)(cyano)methyl)-2-methylphenyl)-2-hydroxybenzamide (ZT-1a' 5 mg/kg per day, IP) or a NF-κB (nuclear factor-κB) inhibitor TAT-NBD (transactivator of transcription-NEMO-binding domain' 20 mg/kg per day, IP). Activation of brain NF-κB and WNK-SPAK-NKCC1 cascade as well as ischemic stroke outcomes were examined. RESULTS Stroke triggered a 2- to 5-fold increase of WNK (isoforms 1, 2, 4), SPAK/OSR1 (oxidative stress-responsive kinase 1), and NKCC1 protein in the ang II-infused hypertensive mouse brains at 24 hours after stroke, which was associated with increased nuclear translocation of phospho-NF-κB protein in the cortical neurons (a Pearson correlation r of 0.77, P<0.005). The upregulation of WNK-SPAK-NKCC1 cascade proteins resulted from increased NF-κB recruitment on Wnk1, Wnk2, Wnk4, Spak, and Nkcc1 gene promoters and was attenuated by NF-κB inhibitor TAT-NBD. Poststroke administration of SPAK inhibitor ZT-1a significantly reduced WNK-SPAK-NKCC1 complex activation, brain lesion size, and neurological function deficits in the ang II-hypertensive mice without affecting blood pressure and cerebral blood flow. CONCLUSIONS The ang II-induced stimulation of NF-κB transcriptional activity upregulates brain WNK-SPAK-NKCC1 cascade and contributes to worsened ischemic stroke outcomes, illustrating the brain WNK-SPAK-NKCC1 complex as a therapeutic target for stroke with comorbid hypertension.
Collapse
Affiliation(s)
- Mohammad Iqbal H Bhuiyan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational, and Clinical Center, PA (M.I.H.B.' D.S.)
| | - Cullen B Young
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Israt Jahan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Md Nabiul Hasan
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Sydney Fischer
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Nur Farah Meor Azlan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom (N.F.M.A., J.Z.)
| | - Mingjun Liu
- Medicine (M.L., D.G.), University of Pittsburgh, PA
| | - Ansuman Chattopadhyay
- Molecular Biology-Information Service, Health Sciences Library System (A.C.), University of Pittsburgh, PA
| | - Huachen Huang
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
| | - Kristopher T Kahle
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston (K.T.K.)
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom (N.F.M.A., J.Z.)
| | | | - Bradley J Molyneaux
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
| | - Adam C Straub
- Pharmacology and Chemical Biology (A.C.S), University of Pittsburgh, PA
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (A.C.S., D.G.), University of Pittsburgh, PA
| | - Xianming Deng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Fujian, China (X.D.)
| | - Delphine Gomez
- Medicine (M.L., D.G.), University of Pittsburgh, PA
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (A.C.S., D.G.), University of Pittsburgh, PA
| | - Dandan Sun
- Departments of Neurology (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., H.H., B.J.M., D.S.), University of Pittsburgh, PA
- Pittsburgh Institute for Neurodegenerative Disorders (M.I.H.B., C.B.Y., I.J., M.N.H., S.F., B.J.M., D.S.), University of Pittsburgh, PA
- Veterans Affairs Pittsburgh Health Care System, Geriatric Research, Educational, and Clinical Center, PA (M.I.H.B.' D.S.)
| |
Collapse
|
16
|
Dietary salt with nitric oxide deficiency induces nocturnal polyuria in mice via hyperactivation of intrarenal angiotensin II-SPAK-NCC pathway. Commun Biol 2022; 5:175. [PMID: 35228649 PMCID: PMC8885931 DOI: 10.1038/s42003-022-03104-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 02/01/2022] [Indexed: 11/12/2022] Open
Abstract
Nocturnal polyuria is the most frequent cause of nocturia, a common disease associated with a compromised quality of life and increased mortality. Its pathogenesis is complex, and the detailed underlying mechanism remains unknown. Herein, we report that concomitant intake of a high-salt diet and reduced nitric oxide (NO) production achieved through Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME) administration in mice resulted in nocturnal polyuria recapitulating the clinical features in humans. High salt intake under reduced NO production overactivated the angiotensin II-SPAK (STE20/SPS1-related proline–alanine-rich protein kinase)-NCC (sodium chloride co-transporter) pathway in the kidney, resulting in the insufficient excretion of sodium during the day and its excessive excretion at night. Excessive Na excretion at night in turn leads to nocturnal polyuria due to osmotic diuresis. Our study identified a central role for the intrarenal angiotensin II-SPAK-NCC pathway in the pathophysiology of nocturnal polyuria, highlighting its potential as a promising therapeutic target. This study reports a mouse model of nocturnal polyuria - increased urine production at night that causes compromised quality of life and may impact mortality in older people. The authors identify a molecular pathway in the kidney that could prove to be a promising drug target for nocturnal polyuria.
Collapse
|
17
|
Prisco SZ, Eklund M, Raveendran R, Thenappan T, Prins KW. With No Lysine Kinase 1 Promotes Metabolic Derangements and RV Dysfunction in Pulmonary Arterial Hypertension. JACC. BASIC TO TRANSLATIONAL SCIENCE 2021; 6:834-850. [PMID: 34869947 PMCID: PMC8617575 DOI: 10.1016/j.jacbts.2021.09.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/31/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022]
Abstract
Small molecule inhibition of with no lysine kinase 1 (WNK1) (WNK463) signaling activates adenosine monophosphate-activated protein kinase signaling and mitigates membrane enrichment of glucose transporters 1 and 4, which decreases protein O-GlcNAcylation and glycation. Quantitative proteomics of right ventricular (RV) mitochondrial enrichments shows WNK463 prevents down-regulation of several mitochondrial metabolic enzymes. and metabolomics analysis suggests multiple metabolic processes are corrected. Physiologically, WNK463 augments RV systolic and diastolic function independent of pulmonary arterial hypertension severity. Hypochloremia, a condition of predicted WNK1 activation in patients with pulmonary arterial hypertension, is associated with more severe RV dysfunction. These results suggest WNK1 may be a druggable target to combat metabolic dysregulation and may improve RV function and survival in pulmonary arterial hypertension.
Collapse
Key Words
- AMPK, adenosine monophosphate-activated protein kinase
- AS160, 160 kDa substrate of the Akt serine/threonine kinase
- DCA, dicarboxylic fatty acid
- FAO, fatty acid oxidation
- GLO1, glyoxalase 1
- GLO2, glyoxalase 2
- GLUT1, glucose transporter 1
- GLUT4, glucose transporter 4
- LV, left ventricle/ventricular
- MCT, monocrotaline
- MCT-V, monocrotaline-vehicle
- PAH, pulmonary arterial hypertension
- PTM, post-translationally modify/modifications
- PV, pressure-volume
- PVR, pulmonary vascular resistance
- RA, right atrial
- RV, right ventricle/ventricular
- RVD, right ventricular dysfunction
- TCA, tricarboxylic acid
- Tau/τ, right ventricular relaxation time
- UDP-GlcNAC, uridine diphosphate N-acetylglucosamine
- WNK, with no lysine kinase
- lipotoxicity
- metabolism
- mitochondria
- pulmonary arterial hypertension
- right ventricular dysfunction
- with no lysine kinase 1
Collapse
Affiliation(s)
| | | | | | | | - Kurt W. Prins
- Address for correspondence: Dr Kurt Prins, Lillehei Heart Institute, Cardiovascular Division, University of Minnesota Medical School, 312 Church Street Southeast, Minneapolis, Minnesota 55455, USA.
| |
Collapse
|
18
|
Radloff J, Latic N, Pfeiffenberger U, Schüler C, Tangermann S, Kenner L, Erben RG. A phosphate and calcium-enriched diet promotes progression of 5/6-nephrectomy-induced chronic kidney disease in C57BL/6 mice. Sci Rep 2021; 11:14868. [PMID: 34290280 PMCID: PMC8295299 DOI: 10.1038/s41598-021-94264-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
C57BL/6 mice are known to be rather resistant to the induction of experimental chronic kidney disease (CKD) by 5/6-nephrectomy (5/6-Nx). Here, we sought to characterize the development of CKD and its cardiac and skeletal sequelae during the first three months after 5/6-Nx in C57BL/6 mice fed a calcium- and phosphate enriched diet (CPD) with a balanced calcium/phosphate ratio. 5/6-NX mice on CPD showed increased renal fibrosis and a more pronounced decrease in glomerular filtration rate when compared to 5/6-Nx mice on normal diet (ND). Interestingly, despite comparable levels of serum calcium, phosphate, and parathyroid hormone (PTH), circulating intact fibroblast growth factor-23 (FGF23) was 5 times higher in 5/6-Nx mice on CPD, relative to 5/6-Nx mice on ND. A time course experiment revealed that 5/6-Nx mice on CPD developed progressive renal functional decline, renal fibrosis, cortical bone loss, impaired bone mineralization as well as hypertension, but not left ventricular hypertrophy. Collectively, our data show that the resistance of C57BL/6 mice to 5/6-Nx can be partially overcome by feeding the CPD, and that the CPD induces a profound, PTH-independent increase in FGF23 in 5/6-Nx mice, making it an interesting tool to assess the pathophysiological significance of FGF23 in CKD.
Collapse
Affiliation(s)
- J Radloff
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - N Latic
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - U Pfeiffenberger
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - C Schüler
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria
| | - S Tangermann
- Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - L Kenner
- Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - R G Erben
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, Veterinaerplatz 1, 1210, Vienna, Austria.
| |
Collapse
|
19
|
Nagatani Y, Higashino T, Kinoshita K, Higashino H. Thromboxane A 2 receptor antagonist (ONO-8809) attenuates renal disorders caused by salt overload in stroke-prone spontaneously hypertensive rats. Clin Exp Pharmacol Physiol 2021; 48:1391-1401. [PMID: 34152603 PMCID: PMC8518871 DOI: 10.1111/1440-1681.13543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/11/2021] [Accepted: 06/18/2021] [Indexed: 11/28/2022]
Abstract
Epidemiological and clinical studies have demonstrated that excessive salt intake causes severe hypertension and exacerbates organ derangement, such as in chronic kidney disease (CKD). In this study, we focused on evaluating the histological and gene expression effects in the kidneys of stroke‐prone spontaneously hypertensive rats (SHRSP) with a high salt intake and the thromboxane A2/ prostaglandin H2 receptor (TPR) blocker ONO‐8809. Six‐week‐old SHRSPs were divided into three groups and were fed normal chow containing 0.4% NaCl, 2.0%NaCl or 2.0%NaCl + ONO‐8809 (0.6 mg/kg p.o. daily). Histological analyses with immunohistochemistry and a gene expression assay with a DNA kidney microarray were performed after 8 weeks. The following changes were observed in SHRSPs with the high salt intake. Glomerular sclerotic changes were remarkably observed in the juxtamedullary cortex areas. The ED1, monocyte chemoattractant protein‐1 (MCP‐1), nitrotyrosine and hypoxia inducible factor 1α (HIF‐1α) staining areas were increased in the glomeruli and interstitial portion of the kidneys. The genes Tbxa2r (that encodes TPR), Prcp and Car7 were significantly underexpressed in the kidneys. The plasma 8‐isoprostane level was significantly elevated and was attenuated with the ONO‐8809 treatment. Thromboxane A2 (TXA2) and oxidative stress exaggerated renal dysfunction in the salt‐loaded SHRSPs, and ONO‐8809 as a TPR blocker suppressed these changes. Therefore, ONO‐8809 is a candidate drug to prevent CKD in hypertensive patients when CKD is associated with a high salt intake.
Collapse
Affiliation(s)
- Yusuke Nagatani
- Department of Pharmacology, Kindai University School of Medicine, Osaka, Japan.,The First Hospital of Welfare-Medical Association, Osaka, Japan
| | - Toshihide Higashino
- Department of Dermatology, Self Defense Forces' Central Hospital, Setagaya, Tokyo, Japan.,Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kosho Kinoshita
- Department of Pharmacology, Kindai University School of Medicine, Osaka, Japan.,Kosho Clinic, Nishinomiya, Hyogo, Japan
| | - Hideaki Higashino
- Department of Pharmacology, Kindai University School of Medicine, Osaka, Japan
| |
Collapse
|
20
|
Nanamatsu A, Mori T, Ando F, Furusho T, Mandai S, Susa K, Sohara E, Rai T, Uchida S. Vasopressin Induces Urinary Uromodulin Secretion By Activating PKA (Protein Kinase A). Hypertension 2021; 77:1953-1963. [PMID: 33896194 DOI: 10.1161/hypertensionaha.121.17127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Azuma Nanamatsu
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Takayasu Mori
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Fumiaki Ando
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Taisuke Furusho
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Shintaro Mandai
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Koichiro Susa
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Tatemitsu Rai
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, (TMDU) Bunkyo, Japan
| |
Collapse
|
21
|
Bovée DM, Uijl E, Severs D, Rubio-Beltrán E, van Veghel R, Maassen van den Brink A, Joles JA, Zietse R, Cuevas CA, Danser AHJ, Hoorn EJ. Dietary salt modifies the blood pressure response to renin-angiotensin inhibition in experimental chronic kidney disease. Am J Physiol Renal Physiol 2021; 320:F654-F668. [PMID: 33586496 DOI: 10.1152/ajprenal.00603.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic kidney disease contributes to hypertension, but the mechanisms are incompletely understood. To address this, we applied the 5/6th nephrectomy rat model to characterize hypertension and the response to dietary salt and renin-angiotensin inhibition. 5/6th nephrectomy caused low-renin, salt-sensitive hypertension with hyperkalemia and unsuppressed aldosterone. Compared with sham rats, 5/6th nephrectomized rats had lower Na+/H+ exchanger isoform 3, Na+-K+-2Cl- cotransporter, Na+-Cl- cotransporter, α-epithelial Na+ channel (ENaC), and Kir4.1 levels but higher serum and glucocorticoid-regulated kinase 1, prostasin, γ-ENaC, and Kir5.1 levels. These differences correlated with plasma renin, aldosterone, and/or K+. On a normal-salt diet, adrenalectomy (0 ± 9 mmHg) and spironolactone (-11 ± 10 mmHg) prevented a progressive rise in blood pressure (10 ± 8 mmHg), and this was enhanced in combination with losartan (-41 ± 12 and -43 ± 9 mmHg). A high-salt diet caused skin Na+ and water accumulation and aggravated hypertension that could only be attenuated by spironolactone (-16 ± 7 mmHg) and in which the additive effect of losartan was lost. Spironolactone also increased natriuresis, reduced skin water accumulation, and restored vasorelaxation. In summary, in the 5/6th nephrectomy rat chronic kidney disease model, salt-sensitive hypertension develops with a selective increase in γ-ENaC and despite appropriate transporter adaptations to low renin and hyperkalemia. With a normal-salt diet, hypertension in 5/6th nephrectomy depends on angiotensin II and aldosterone, whereas a high-salt diet causes more severe hypertension mediated through the mineralocorticoid receptor.NEW & NOTEWORTHY Chronic kidney disease (CKD) causes salt-sensitive hypertension, but the interactions between dietary salt and the renin-angiotensin system are incompletely understood. In rats with CKD on a normal-salt diet targeting aldosterone, the mineralocorticoid receptor (MR) and especially angiotensin II reduced blood pressure. On a high-salt diet, however, only MR blockade attenuated hypertension. These results reiterate the importance of dietary salt restriction to maintain renin-angiotensin system inhibitor efficacy and specify the MR as a target in CKD.
Collapse
Affiliation(s)
- Dominique M Bovée
- Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Estrellita Uijl
- Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - David Severs
- Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eloisa Rubio-Beltrán
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Richard van Veghel
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Antoinette Maassen van den Brink
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Robert Zietse
- Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Catherina A Cuevas
- Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - A H Jan Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ewout J Hoorn
- Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
22
|
WNK1-TAK1 signaling suppresses lipopolysaccharide-induced cytokine production and classical activation in macrophages. Biochem Biophys Res Commun 2020; 533:1290-1297. [PMID: 33046244 DOI: 10.1016/j.bbrc.2020.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 12/17/2022]
Abstract
With-no-lysine kinase (WNK) plays important roles in regulating electrolyte homeostasis, cell signaling, survival, and proliferation. It has been recently demonstrated that WNK1, a member of the WNK family, modifies the function of immune cells. Here we report that in macrophages, WNK1 has suppressive effects on lipopolysaccharide (LPS)-induced inflammatory responses via TGFβ-activated kinase 1 (TAK1)-mediated activation of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathway. We found that WNK1 heterozygous (WNK1+/-) mice produced excessive proinflammatory cytokines in an experimental LPS-induced sepsis model, and peritoneal macrophages isolated from WNK1+/- mice produced higher levels of LPS-induced cytokines and NOS2 expression as canonical proinflammatory M1 macrophage markers. We confirmed that small hairpin RNA (shRNA)-mediated knockdown of WNK1 activated LPS-induced cytokine production and NOS2 expression in RAW 264.7 macrophages. Moreover, we demonstrated that WNK1 knockdown increased the nuclear translocation of NF-κB and activated the p38 and Jun N-terminal kinase (JNK) MAPK signaling pathway and that a TAK1 inhibitor diminished these effects of WNK1 knockdown. These results suggest that WNK1 acts as a physiologic immune modulator via interactions with TAK1. WNK1 may be a therapeutic target against the cytokine storm caused by sepsis.
Collapse
|
23
|
Bovée DM, Cuevas CA, Zietse R, Danser AHJ, Mirabito Colafella KM, Hoorn EJ. Salt-sensitive hypertension in chronic kidney disease: distal tubular mechanisms. Am J Physiol Renal Physiol 2020; 319:F729-F745. [DOI: 10.1152/ajprenal.00407.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic kidney disease (CKD) causes salt-sensitive hypertension that is often resistant to treatment and contributes to the progression of kidney injury and cardiovascular disease. A better understanding of the mechanisms contributing to salt-sensitive hypertension in CKD is essential to improve these outcomes. This review critically explores these mechanisms by focusing on how CKD affects distal nephron Na+ reabsorption. CKD causes glomerulotubular imbalance with reduced proximal Na+ reabsorption and increased distal Na+ delivery and reabsorption. Aldosterone secretion further contributes to distal Na+ reabsorption in CKD and is not only mediated by renin and K+ but also by metabolic acidosis, endothelin-1, and vasopressin. CKD also activates the intrarenal renin-angiotensin system, generating intratubular angiotensin II to promote distal Na+ reabsorption. High dietary Na+ intake in CKD contributes to Na+ retention by aldosterone-independent activation of the mineralocorticoid receptor mediated through Rac1. High dietary Na+ also produces an inflammatory response mediated by T helper 17 cells and cytokines increasing distal Na+ transport. CKD is often accompanied by proteinuria, which contains plasmin capable of activating the epithelial Na+ channel. Thus, CKD causes both local and systemic changes that together promote distal nephron Na+ reabsorption and salt-sensitive hypertension. Future studies should address remaining knowledge gaps, including the relative contribution of each mechanism, the influence of sex, differences between stages and etiologies of CKD, and the clinical relevance of experimentally identified mechanisms. Several pathways offer opportunities for intervention, including with dietary Na+ reduction, distal diuretics, renin-angiotensin system inhibitors, mineralocorticoid receptor antagonists, and K+ or H+ binders.
Collapse
Affiliation(s)
- Dominique M. Bovée
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Catharina A. Cuevas
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Robert Zietse
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A. H. Jan Danser
- Division of Vascular Medicine, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Katrina M. Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Ewout J. Hoorn
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Yamazaki O, Hirohama D, Ishizawa K, Shibata S. Role of the Ubiquitin Proteasome System in the Regulation of Blood Pressure: A Review. Int J Mol Sci 2020; 21:E5358. [PMID: 32731518 PMCID: PMC7432568 DOI: 10.3390/ijms21155358] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/24/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
The kidney and the vasculature play crucial roles in regulating blood pressure. The ubiquitin proteasome system (UPS), a multienzyme process mediating covalent conjugation of the 76-amino acid polypeptide ubiquitin to a substrate protein followed by proteasomal degradation, is involved in multiple cellular processes by regulating protein turnover in various tissues. Increasing evidence demonstrates the roles of UPS in blood pressure regulation. In the kidney, filtered sodium is reabsorbed through diverse sodium transporters and channels along renal tubules, and studies conducted till date have provided insights into the complex molecular network through which ubiquitin ligases modulate sodium transport in different segments. Components of these pathways include ubiquitin ligase neuronal precursor cell-expressed developmentally downregulated 4-2, Cullin-3, and Kelch-like 3. Moreover, accumulating data indicate the roles of UPS in blood vessels, where it modulates nitric oxide bioavailability and vasoconstriction. Cullin-3 not only regulates renal salt reabsorption but also controls vascular tone using different adaptor proteins that target distinct substrates in vascular smooth muscle cells. In endothelial cells, UPS can also contribute to blood pressure regulation by modulating endothelial nitric oxide synthase. In this review, we summarize current knowledge regarding the role of UPS in blood pressure regulation, focusing on renal sodium reabsorption and vascular function.
Collapse
Affiliation(s)
| | | | | | - Shigeru Shibata
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8605, Japan; (O.Y.); (D.H.); (K.I.)
| |
Collapse
|
25
|
Furusho T, Uchida S, Sohara E. The WNK signaling pathway and salt-sensitive hypertension. Hypertens Res 2020; 43:733-743. [PMID: 32286498 DOI: 10.1038/s41440-020-0437-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/19/2022]
Abstract
The distal nephron of the kidney has a central role in sodium and fluid homeostasis, and disruption of this homeostasis due to mutations of with-no-lysine kinase 1 (WNK1), WNK4, Kelch-like 3 (KLHL3), or Cullin 3 (CUL3) causes pseudohypoaldosteronism type II (PHAII), an inherited hypertensive disease. WNK1 and WNK4 activate the NaCl cotransporter (NCC) at the distal convoluted tubule through oxidative stress-responsive gene 1 (OSR1)/Ste20-related proline-alanine-rich kinase (SPAK), constituting the WNK-OSR1/SPAK-NCC phosphorylation cascade. The level of WNK protein is regulated through degradation by the CUL3-KLHL3 E3 ligase complex. In the normal state, the activity of WNK signaling in the kidney is physiologically regulated by sodium intake to maintain sodium homeostasis in the body. In patients with PHAII, however, because of the defective degradation of WNK kinases, NCC is constitutively active and not properly suppressed by a high salt diet, leading to abnormally increased salt reabsorption and salt-sensitive hypertension. Importantly, recent studies have demonstrated that potassium intake, insulin, and TNFα are also physiological regulators of WNK signaling, suggesting that they contribute to the salt-sensitive hypertension associated with a low potassium diet, metabolic syndrome, and chronic kidney disease, respectively. Moreover, emerging evidence suggests that WNK signaling also has some unique roles in metabolic, cardiovascular, and immunological organs. Here, we review the recent literature and discuss the molecular mechanisms of the WNK signaling pathway and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Taisuke Furusho
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
26
|
Wen Y, Crowley SD. Connecting cytokines and cellular signals in the nephron during CKD and hypertension. Kidney Int 2020; 97:651-653. [PMID: 32200858 DOI: 10.1016/j.kint.2020.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/12/2020] [Accepted: 01/13/2020] [Indexed: 10/24/2022]
Abstract
Chronic kidney disease features chronic inflammation and fibrosis, both of which contribute to and are exacerbated by arterial hypertension. The contribution of immune responses to renal sodium retention has received intense scrutiny. In this regard, the article by Furusho et al. details a mechanism wherein intrarenal TNFα augments salt-sensitive hypertension during CKD via activation of the WNK1-SPAKNCC phosphorylation cascade.
Collapse
Affiliation(s)
- Yi Wen
- Division of Nephrology, Department of Medicine, Duke University, Durham, North Carolina, USA; Department of Medicine, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University, Durham, North Carolina, USA; Department of Medicine, Durham Veterans Affairs Medical Center, Durham, North Carolina, USA.
| |
Collapse
|