1
|
Sheu JY, Chang LY, Chen JY, Chuang MH, Wu VC, Chueh JS. The Prevalence of Obstructive Sleep Apnea in Patients With Primary Aldosteronism. J Clin Endocrinol Metab 2024; 109:2681-2691. [PMID: 38941133 DOI: 10.1210/clinem/dgae415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Indexed: 06/29/2024]
Abstract
CONTEXT Investigating the co-occurrence of obstructive sleep apnea (OSA) and primary aldosteronism (PA) is crucial for understanding their interrelation. OBJECTIVE This work aimed to evaluate the prevalence of OSA in individuals diagnosed with PA and to assess the prevalence of PA within the OSA population, with a specific focus on hypertensive individuals. METHODS An exhaustive search was performed across PubMed, Embase, CINAHL, Scopus, and Web of Science up to September 2023, without restrictions on language or publication date. Studies were selected based on their focus on the prevalence of OSA in PA patients and vice versa, specifically in hypertensive individuals. Data were extracted using standard guidelines, focusing on patient characteristics, prevalence rates, and other relevant clinical parameters. RESULTS Proportional meta-analysis using a random-effects model revealed a 59.8% prevalence of OSA in hypertensive PA patients, with 45.4% exhibiting moderate-to-severe OSA. Meta-regression showed no significant effect of age, sex, body mass index, antihypertensive medication, systolic blood pressure, diastolic blood pressure, or serum potassium on OSA prevalence. However, a significant positive association was found with the glomerular filtration rate (GFR) (P < .001). Subgroup analysis also revealed that a hyperfiltration rate (GFR ≥ 100 mL/min per 1.73 m2) may be associated with a higher prevalence of OSA (71%, P value for interaction < .01). Among hypertensive OSA patients, 11.2% had PA. CONCLUSION A substantial prevalence of OSA in individuals with PA was identified, demonstrating a complex interplay between these conditions in hypertensive patients. Notably, the prevalence of OSA was significantly associated with kidney hyperfiltration.
Collapse
Affiliation(s)
- Jia-Yuh Sheu
- Department of Urology, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Li-Yang Chang
- College of Medicine, National Taiwan University, Taipei 106319, Taiwan
| | - Jui-Yi Chen
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan 71004, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Min-Hsiang Chuang
- Division of Nephrology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
- Primary Aldosteronism Center of National Taiwan University Hospital, Taipei 100225, Taiwan
- Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Jeff S Chueh
- Department of Urology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Primary Aldosteronism Center of National Taiwan University Hospital, Taipei 100225, Taiwan
- Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan
| |
Collapse
|
2
|
Rendell M. Lessons learned from early-stage clinical trials for diabetic nephropathy. Expert Opin Investig Drugs 2024; 33:287-301. [PMID: 38465470 DOI: 10.1080/13543784.2024.2326025] [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: 08/13/2023] [Accepted: 02/28/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION The evolution of treatment for diabetic nephropathy illustrates how basic biochemistry and physiology have led to new agents such as SGLT2 inhibitors and mineralocorticoid blockers. Conversely, clinical studies performed with these agents have suggested new concepts for investigational drug development. We reviewed currently available treatments for diabetic nephropathy and then analyzed early clinical trials of new agents to assess the potential for future treatment modalities. AREAS COVERED We searched ClinicalTrials.gov for new agents under study for diabetic nephropathy in the past decade. Once we have identified investigation trials of new agents, we then used search engines and Pubmed.gov to find publications providing insight on these drugs. Current treatments have shown benefit in both cardiac and renal disease. In our review, we found 51 trials and 43 pharmaceuticals in a number of drug classes: mineralocorticoid blockers, anti-inflammatory, anti-fibrosis, nitric oxide stimulatory, and podocyte protection, and endothelin inhibitors. EXPERT OPINION It is difficult to predict which early phase treatments will advance to confirmatory clinical trials. Current agents are thought to improve hemodynamic function. However, the coincident benefit of both myocardial function and the glomerulus argues for primary effects at the subcellular level, and we follow the evolution of agents which modify fundamental cellular processes.
Collapse
Affiliation(s)
- Marc Rendell
- The Association of Diabetes Investigators, Newport Coast, CA, USA
- The Rose Salter Medical Research Foundation, Newport Coast, CA, USA
| |
Collapse
|
3
|
Bayne S, LeFevre J, Olstinske K, Ravindran S, Munusamy S. Renoprotective Effects of Mineralocorticoid Receptor Antagonists Against Diabetic Kidney Disease. Adv Biol (Weinh) 2024; 8:e2300496. [PMID: 38065929 DOI: 10.1002/adbi.202300496] [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: 09/13/2023] [Revised: 10/18/2023] [Indexed: 03/16/2024]
Abstract
Diabetic kidney disease (DKD) is a growing epidemic worldwide and a leading cause of end-stage kidney disease. Mineralocorticoid receptor (MR) blockade using Finerenone is a recently approved therapeutic approach to slow down the progression of DKD in patients with type 2 diabetes in addition to other therapies such as angiotensin-II converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), sodium-glucose co-transporter 2 (SGLT2) inhibitors, and glucagon-like peptide 1 (GLP-1) analogs. This review elaborates on the pathophysiologic pathways activated by aldosterone (the human mineralocorticoid) in DKD, the pharmacology of three different generations of mineralocorticoid receptor antagonists (MRAs), specifically, spironolactone, eplerenone, and finerenone, and the mechanisms by which these MRAs elicit their protective effects on the kidney under diabetic settings.
Collapse
Affiliation(s)
- Sarah Bayne
- Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| | - James LeFevre
- Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| | - Kayla Olstinske
- Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| | | | - Shankar Munusamy
- Department of Pharmaceutical and Administrative Sciences, Drake University College of Pharmacy and Health Sciences, Des Moines, IA, 50311, USA
| |
Collapse
|
4
|
Sheu JY, Wu VC, Chueh JS. Re: comment from Ladak et al. to JH-D-23-00692, entitled 'Analyzing eGFR-Dip after medical target therapy in primary aldosteronism'. J Hypertens 2024; 42:377-378. [PMID: 38165052 DOI: 10.1097/hjh.0000000000003611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Affiliation(s)
- Jia-Yuh Sheu
- Department of Urology, National Taiwan University Hospital
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taiwan
| | - Jeff S Chueh
- Department of Urology, National Taiwan University Hospital
| |
Collapse
|
5
|
Lai CF, Lin YH, Huang KH, Chueh JS, Wu VC. Kidney function predicts new-onset cardiorenal events and mortality in primary aldosteronism: approach of the 2021 race-free eGFR equation. Hypertens Res 2024; 47:233-244. [PMID: 37714953 DOI: 10.1038/s41440-023-01400-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 09/17/2023]
Abstract
Individuals with primary aldosteronism (PA) exhibit glomerular hyperfiltration, which may conceal underlying kidney damage. This observational cohort study enrolled 760 coronary artery disease-naive patients diagnosed with PA between January 1, 2007 and December 31, 2018 (male, 45%; mean age, 52.3 ± 11.9 years). The baseline estimated glomerular filtration rate (eGFR) was calculated using the 2021 Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation, which includes serum creatinine and cystatin C but omits the race variable. During a mean follow-up of 5.8 ± 3.2 years, new-onset composite cardiovascular events (total death, non-fatal myocardial infarction, and coronary revascularization procedure) occurred at a crude incidence rate of 10.9 per 1,000 person-years. Multivariable Cox proportional hazards analysis showed that baseline eGFR was independently associated with composite cardiovascular events (hazard ratio [HR], 0.98 [95% CI, 0.97-0.99]). Penalized splines smoothing in multivariable regression analysis demonstrated that the risk of composite cardiovascular events increased negatively and linearly when patients had a baseline eGFR less than 85 mL/min/1.73 m2. Patients with baseline eGFR <85 mL/min/1.73 m2 were independently associated with higher risks of composite cardiovascular events (HR, 2.39 [95% CI, 1.16-4.93]), all-cause mortality (HR, 4.63 [95% CI, 1.59-13.46]), and adverse kidney events (sub-distribution HR, 5.96 [95% CI, 3.69-9.62], with mortality as a competing risk). Our data support baseline eGFR as a predictor for new-onset adverse cardiorenal events and emphasizes the importance of the early detection of kidney function impairment in hypertensive patients with PA. We also firstly validate the 2021 race-free CKD-EPI eGFR equation in Asian patents with PA. Even with the glomerular hyperfiltration phenomenon, baseline eGFR in patients with primary aldosteronism is associated with subsequent cardiorenal outcomes. The results also firstly point to the validity of the 2021 race-free CKD-EPI eGFR equation in healthcare and clinical decision-making.
Collapse
Affiliation(s)
- Chun-Fu Lai
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Primary Aldosteronism Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-How Huang
- Department of Urology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jeff S Chueh
- Department of Urology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Vin-Cent Wu
- Renal Division, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
- Primary Aldosteronism Center, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
6
|
Yoshida Y, Shibata H. No "U-shaped" associations of estimated glomerular filtration rate with adverse cardiovascular outcomes in patients with primary aldosteronism. Hypertens Res 2024; 47:247-249. [PMID: 37945892 DOI: 10.1038/s41440-023-01506-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Affiliation(s)
- Yuichi Yoshida
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, Yufu, Japan.
| |
Collapse
|
7
|
Sheu JY, Wang SM, Wu VC, Huang KH, Tseng CS, Lee YJ, Tsai YC, Lin YH, Chueh JS. Estimated glomerular filtration rate-dip after medical target therapy associated with increased mortality and cardiovascular events in patients with primary aldosteronism. J Hypertens 2023; 41:1401-1410. [PMID: 37334546 DOI: 10.1097/hjh.0000000000003479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
OBJECTIVES The correlation of the changes of estimated glomerular filtration rate (eGFR) with long-term cardiovascular complications in patients with primary aldosteronism (PA) following mineralocorticoid receptor antagonists (MRA) treatment remains ambiguous. This prospective study aims to determine factors associated with all-cause mortality and de novo cardiovascular events of PA patients against the eGFR-dip. METHODS A total of 208 newly diagnosed PA patients were enrolled from January 2017 to January 2019. MRA was administered with at least a 6-month follow-up. The 'eGFR-dip' was defined as the difference between eGFR at 6 months after MRA treatment and respective baseline eGFR divided by the baseline eGFR. RESULTS After a mean 5.7 years follow-up, an eGFR-dip more than 12%, which was detected in 99 (47.6%) of the 208 patients, was a significant independent risk factor predicting composite outcomes (all-cause mortality, de-novo three-point major adverse cardiovascular events, and/or congestive heart failure). Multivariable logistic regression showed that age [odds ratio (OR), 0.94; P = 0.003], pretreatment plasma aldosterone concentration (PAC; OR, 0.98; P = 0.004), and initial eGFR (OR, 0.97; P < 0.001) had a positive linkage with the eGFR-dip more than 12%. CONCLUSIONS Nearly half of PA patients had an eGFR-dip more than 12% after 6 months of MRA treatment. They had a higher incidence of all-cause mortality and de novo cardiovascular events. Elder age, higher pretreatment PAC, or higher initial eGFR could be associated with an elevated risk of an eGFR-dip more than 12%.
Collapse
Affiliation(s)
- Jia-Yuh Sheu
- Department of Medical Education, National Taiwan University Hospital
| | - Shuo-Meng Wang
- Department of Urology, National Taiwan University Hospital
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group
- Primary Aldosteronism Center of National Taiwan University Hospital, Taipei, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group
- Primary Aldosteronism Center of National Taiwan University Hospital, Taipei, Taiwan
| | - Kuo-How Huang
- Department of Urology, National Taiwan University Hospital
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group
- Primary Aldosteronism Center of National Taiwan University Hospital, Taipei, Taiwan
| | - Chi-Shin Tseng
- Department of Urology, National Taiwan University Hospital
| | - Yuan-Ju Lee
- Department of Urology, National Taiwan University Hospital
| | - Yao-Chou Tsai
- Division of Urology, Department of Surgery, Taipei Tzuchi Hospital, The Buddhist Tzu Chi Medical Foundation, New Taipei City
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group
- Primary Aldosteronism Center of National Taiwan University Hospital, Taipei, Taiwan
| | - Jeff S Chueh
- Department of Urology, National Taiwan University Hospital
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group
- Primary Aldosteronism Center of National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
8
|
Sutovska H, Molcan L, Majzunova M, Sykora M, Kopkan L, Zeman M. Mineralocorticoid receptor blockade protects the kidneys but does not affect inverted blood pressure rhythm in hypertensive transgenic (mRen-2)27 rats. Mol Cell Endocrinol 2023; 572:111967. [PMID: 37210027 DOI: 10.1016/j.mce.2023.111967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/17/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
Aldosterone regulates blood pressure (BP) through water and sodium balance. In our study, we studied if continuous treatment with a mineralocorticoid receptor antagonist, spironolactone (30 mg/kg/day) for 20 days can: 1) attenuate hypertension development and restore inverted 24-h BP rhythm in hypertensive transgenic (mRen-2)27 rats (TGR) measured by telemetry; 2) improve function of the kidneys and heart; 3) be protective against high salt load (1% in water) by mitigating oxidative injury and improving kidney function. Spironolactone decreased albuminuria and 8-isoprostane in normal and salt load conditions in BP-independent effects. Salt load increased BP, impaired autonomic balance, suppressed plasma aldosterone level and increased natriuresis, albuminuria and oxidative injury in TGR. Spironolactone did not restore the inverted 24-h rhythm of BP in TGR, therefore, mineralocorticoids are not crucial in regulation of BP daily profile. Spironolactone improved kidney function, decreased oxidative stress and was protective against high salt load in the BP-independent manner.
Collapse
Affiliation(s)
- Hana Sutovska
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic.
| | - Lubos Molcan
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic.
| | - Miroslava Majzunova
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic.
| | - Matus Sykora
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | - Libor Kopkan
- Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Michal Zeman
- Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic.
| |
Collapse
|
9
|
Wu VC, Chan CK, Chueh JS, Chen YM, Lin YH, Chang CC, Lin PC, Chung SD. Markers of Kidney Tubular Function Deteriorate While Those of Kidney Tubule Health Improve in Primary Aldosteronism After Targeted Treatments. J Am Heart Assoc 2023; 12:e028146. [PMID: 36789834 PMCID: PMC10111488 DOI: 10.1161/jaha.122.028146] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Background Targeted treatment with mineralocorticoid receptor antagonists (MRAs) or adrenalectomy in patients with primary aldosteronism (PA) causes a decline in estimated glomerular filtration rate; however, the associated simultaneous changes in biomarkers of kidney tubule health still remain unclear. Methods and Results We matched 104 patients with newly diagnosed unilateral PA who underwent adrenalectomy with 104 patients with unilateral PA who were treated with MRAs, 104 patients with bilateral PA treated with MRAs, and 104 patients with essential hypertension who served as controls. Functional biomarkers were measured before the targeted treatment and 1 year after treatment, including serum markers of kidney function (cystatin C, creatinine), urinary markers of proximal renal tubular damage (L-FABP [liver-type fatty-acid binding protein], KIM-1 [kidney injury molecule-1]), serum markers of kidney tubular reserve and mineral metabolism (intact parathyroid hormone), and proteinuria. Compared with the patients with essential hypertension, the patients with PA had higher pretreatment serum intact parathyroid hormone and urinary creatinine-corrected parameters, including L-FABP, KIM-1, and albumin. The patients with essential hypertension and with PA had similar cystatin C levels. After treatment with MRAs or adrenalectomy of unilateral PA and MRAs of bilateral PA, the patients with PA had increased serum cystatin C and decreased urinary L-FABP/creatinine, KIM-1/creatinine, creatinine-based estimated glomerular filtration rate, intact parathyroid hormone, and proteinuria (all P<0.05). In multivariable regression models, a higher urinary L-FABP/creatinine ratio and older age were significantly correlated with the occurrence of kidney failure (estimated glomerular filtration rate dip ≥30%) in the patients with PA after targeted treatment. Conclusions Compared with the matched patients with essential hypertension, the incident patients with PA at diagnosis had higher levels of several biomarkers, including markers of kidney damage, tubular reserve/mineral metabolism, and proteinuria. Functional kidney failure in the patients with PA after treatment could be predicted by a higher baseline urinary L-FABP/creatinine ratio and older age. After targeted treatments in the patients with bilateral or unilateral PA, these biomarkers of kidney tubule health were restored, but creatinine-based estimated glomerular filtration rate declined, which may therefore reflect hemodynamic changes rather than intrinsic damage to kidney tubular cells.
Collapse
Affiliation(s)
- Vin-Cent Wu
- Department of Internal Medicine National Taiwan University Hospital and College of Medicine, National Taiwan University Taipei Taiwan.,TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group Taipei Taiwan.,Primary Aldosteronism Center at National Taiwan University Hospital (NTUH-PAC) Taipei Taiwan
| | - Chieh-Kai Chan
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group Taipei Taiwan.,Department of Internal Medicine National Taiwan University Hospital Hsin-Chu Branch Hsin-Chu County Taiwan
| | - Jeff S Chueh
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group Taipei Taiwan.,Department of Urology National Taiwan University Hospital and College of Medicine, National Taiwan University Taipei Taiwan
| | - Yung-Ming Chen
- Department of Internal Medicine National Taiwan University Hospital and College of Medicine, National Taiwan University Taipei Taiwan
| | - Yen-Hung Lin
- Department of Internal Medicine National Taiwan University Hospital and College of Medicine, National Taiwan University Taipei Taiwan.,TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group Taipei Taiwan.,Primary Aldosteronism Center at National Taiwan University Hospital (NTUH-PAC) Taipei Taiwan
| | - Chin-Chen Chang
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group Taipei Taiwan.,Department of Imaging Medicine National Taiwan University Hospital and College of Medicine, National Taiwan University Taipei Taiwan
| | - Po-Chih Lin
- Department of Internal Medicine National Taiwan University Hospital and College of Medicine, National Taiwan University Taipei Taiwan
| | - Shiu-Dong Chung
- Division of Urology, Department of Surgery Far Eastern Memorial Hospital New Taipei City Taiwan.,Department of Nursing College of Healthcare and Management, General Education Center, Asia Eastern University of Science and Technology New Taipei City Taiwan
| | -
- TAIPAI, Taiwan Primary Aldosteronism Investigation (TAIPAI) Study Group Taipei Taiwan
| |
Collapse
|
10
|
Martínez-Montoro JI, Morales E, Cornejo-Pareja I, Tinahones FJ, Fernández-García JC. Obesity-related glomerulopathy: Current approaches and future perspectives. Obes Rev 2022; 23:e13450. [PMID: 35362662 PMCID: PMC9286698 DOI: 10.1111/obr.13450] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 12/20/2022]
Abstract
Obesity-related glomerulopathy (ORG) is a silent comorbidity which is increasing in incidence as the obesity epidemic escalates. ORG is associated with serious health consequences including chronic kidney disease, end-stage renal disease (ESRD), and increased mortality. Although the pathogenic mechanisms involved in the development of ORG are not fully understood, glomerular hemodynamic changes, renin-angiotensin-aldosterone system (RAAS) overactivation, insulin-resistance, inflammation and ectopic lipid accumulation seem to play a major role. Despite albuminuria being commonly used for the non-invasive evaluation of ORG, promising biomarkers of early kidney injury that are emerging, as well as new approaches with proteomics and metabolomics, might permit an earlier diagnosis of this disease. In addition, the assessment of ectopic kidney fat by renal imaging could be a useful tool to detect and evaluate the progression of ORG. Weight loss interventions appear to be effective in ORG, although large-scale trials are needed. RAAS blockade has a renoprotective effect in patients with ORG, but even so, a significant proportion of patients with ORG will eventually progress to ESRD despite therapeutic efforts. It is noteworthy that certain antidiabetic agents such as sodium-glucose cotransporter 2 inhibitors (SGLT2i) or glucagon-like peptide-1 receptor agonists (GLP-1 RAs) could be useful in the treatment of ORG through different pleiotropic effects. In this article, we review current approaches and future perspectives in the care and treatment of ORG.
Collapse
Affiliation(s)
- José Ignacio Martínez-Montoro
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain.,Faculty of Medicine, University of Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Enrique Morales
- Department of Nephrology, 12 de Octubre University Hospital, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.,Department of Medicine, Complutense University, Madrid, Spain
| | - Isabel Cornejo-Pareja
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain.,Faculty of Medicine, University of Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.,Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco J Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, Málaga, Spain.,Faculty of Medicine, University of Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.,Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - José Carlos Fernández-García
- Faculty of Medicine, University of Málaga, Málaga, Spain.,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.,Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain.,Department of Endocrinology and Nutrition, Regional University Hospital of Málaga, Málaga, Spain
| |
Collapse
|
11
|
Nakamura T, Bonnard B, Palacios-Ramirez R, Fernández-Celis A, Jaisser F, López-Andrés N. Biglycan Is a Novel Mineralocorticoid Receptor Target Involved in Aldosterone/Salt-Induced Glomerular Injury. Int J Mol Sci 2022; 23:ijms23126680. [PMID: 35743123 PMCID: PMC9224513 DOI: 10.3390/ijms23126680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 11/16/2022] Open
Abstract
The beneficial effects of mineralocorticoid receptor (MR) antagonists (MRAs) for various kidney diseases are established. However, the underlying mechanisms of kidney injury induced by MR activation remain to be elucidated. We recently reported aldosterone-induced enhancement of proteoglycan expression in mitral valve interstitial cells and its association with fibromyxomatous valvular disorder. As the expression of certain proteoglycans is elevated in several kidney diseases, we hypothesized that proteoglycans mediate kidney injury in the context of aldosterone/MR pathway activation. We evaluated the proteoglycan expression and tissue injury in the kidney and isolated glomeruli of uninephrectomy/aldosterone/salt (NAS) mice. The MRA eplerenone was administered to assess the role of the MR pathway. We investigated the direct effects of biglycan, one of the proteoglycans, on macrophages using isolated macrophages. The kidney samples from NAS-treated mice showed enhanced fibrosis and increased expression of biglycan accompanying glomerular macrophage infiltration and enhanced expression of TNF-α, iNOS, Nox2, CCL3 (C-C motif chemokine ligand 3), and phosphorylated NF-κB. Eplerenone blunted these changes. Purified biglycan stimulated macrophages to express TNF-α, iNOS, Nox2, and CCL3. This was prevented by a toll-like receptor 4 (TLR4) or NF-κB inhibitor, indicating that biglycan stimulation is dependent on the TLR4/NF-κB pathway. We identified the proteoglycan biglycan as a novel target of MR involved in MR-induced glomerular injury and macrophage infiltration via a biglycan/TLR4/NF-κB/CCL3 cascade.
Collapse
Affiliation(s)
- Toshifumi Nakamura
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
| | - Benjamin Bonnard
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
| | - Roberto Palacios-Ramirez
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (T.N.); (B.B.); (R.P.-R.)
- INSERM, Clinical Investigation Centre 1433, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT (Cardiovascular and Renal Clinical Trialists), 54500 Nancy, France
- Correspondence: (F.J.); (N.L.-A.); Tel.: +33-144276485 (F.J.); +34-848428539 (N.L.-A.)
| | - Natalia López-Andrés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain;
- Correspondence: (F.J.); (N.L.-A.); Tel.: +33-144276485 (F.J.); +34-848428539 (N.L.-A.)
| |
Collapse
|
12
|
Kidney Damage Caused by Obesity and Its Feasible Treatment Drugs. Int J Mol Sci 2022; 23:ijms23020747. [PMID: 35054932 PMCID: PMC8775419 DOI: 10.3390/ijms23020747] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 02/07/2023] Open
Abstract
The rapid growth of obesity worldwide has made it a major health problem, while the dramatic increase in the prevalence of obesity has had a significant impact on the magnitude of chronic kidney disease (CKD), especially in developing countries. A vast amount of researchers have reported a strong relationship between obesity and chronic kidney disease, and obesity can serve as an independent risk factor for kidney disease. The histological changes of kidneys in obesity-induced renal injury include glomerular or tubular hypertrophy, focal segmental glomerulosclerosis or bulbous sclerosis. Furthermore, inflammation, renal hemodynamic changes, insulin resistance and lipid metabolism disorders are all involved in the development and progression of obesity-induced nephropathy. However, there is no targeted treatment for obesity-related kidney disease. In this review, RAS inhibitors, SGLT2 inhibitors and melatonin would be presented to treat obesity-induced kidney injury. Furthermore, we concluded that melatonin can protect the kidney damage caused by obesity by inhibiting inflammation and oxidative stress, revealing its therapeutic potential.
Collapse
|
13
|
Erraez S, López-Mesa M, Gómez-Fernández P. Mineralcorticoid receptor blockers in chronic kidney disease. Nefrologia 2021; 41:258-275. [PMID: 36166243 DOI: 10.1016/j.nefroe.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/17/2020] [Indexed: 06/16/2023] Open
Abstract
There are many experimental data supporting the involvement of aldosterone and mineralcorticoid receptor (MR) activation in the genesis and progression of chronic kidney disease (CKD) and cardiovascular damage. Many studies have shown that in diabetic and non-diabetic CKD, blocking the renin-angiotensin-aldosterone (RAAS) system with conversion enzyme inhibitors (ACEi) or angiotensin II receptor blockers (ARBs) decreases proteinuria, progression of CKD and mortality, but there is still a significant residual risk of developing these events. In subjects treated with ACEi or ARBs there may be an aldosterone breakthrough whose prevalence in subjects with CKD can reach 50%. Several studies have shown that in CKD, the aldosterone antagonists (spironolactone, eplerenone) added to ACEi or ARBs, reduce proteinuria, but increase the risk of hyperkalemia. Other studies in subjects treated with dialysis suggest a possible beneficial effect of antialdosteronic drugs on CV events and mortality. Newer potassium binders drugs can prevent/decrease hyperkalemia induced by RAAS blockade, and may reduce the high discontinuation rates or dose reduction of RAAS-blockers. The nonsteroidal MR blockers, with more potency and selectivity than the classic ones, reduce proteinuria and have a lower risk of hyperkalemia. Several clinical trials, currently underway, will determine the effect of classic MR blockers on CV events and mortality in subjects with stage 3b CKD and in dialysis patients, and whether in patients with type 2 diabetes mellitus and CKD, optimally treated and with high risk of CV and kidney events, the addition of finerenone to their treatment produces cardiorenal benefits. Large randomized trials have shown that sodium glucose type 2 cotransporter inhibitors (SGLT2i) reduce mortality and the development and progression of diabetic and nondiabetic CKD. There are pathophysiological arguments, which raise the possibility that the triple combination ACEi or ARBs, SGLT2i and aldosterone antagonist provide additional renal and cardiovascular protection.
Collapse
Affiliation(s)
- Sara Erraez
- Unidad de Factores de Riesgo Vascular, Nefrología, Hospital Universitario de Jerez, Jerez de la Frontera, Cádiz, Spain
| | | | - Pablo Gómez-Fernández
- Unidad de Factores de Riesgo Vascular, Nefrología, Hospital Universitario de Jerez, Jerez de la Frontera, Cádiz, Spain.
| |
Collapse
|
14
|
[Mineralcorticoid receptor blockers in chronic kidney disease]. Nefrologia 2020; 41:258-275. [PMID: 33358451 DOI: 10.1016/j.nefro.2020.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/17/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022] Open
Abstract
There are many experimental data supporting the involvement of aldosterone and mineralcorticoid receptor (MR) activation in the genesis and progression of chronic kidney disease (CKD) and cardiovascular damage. Many studies have shown that in diabetic and non-diabetic CKD, blocking the renin- angiotensin-aldosterone (RAAS) system with conversion enzyme inhibitors (ACEi) or angiotensin II receptor blockers (ARBs) decreases proteinuria, progression of CKD and mortality, but there is still a significant residual risk of developing these events. In subjects treated with ACEi or ARBs there may be an aldosterone breakthrough whose prevalence in subjects with CKD can reach 50%. Several studies have shown that in CKD, the aldosterone antagonists (spironolactone, eplerenone) added to ACEi or ARBs, reduce proteinuria, but increase the risk of hyperkalemia. Other studies in subjects treated with dialysis suggest a possible beneficial effect of antialdosteronic drugs on CV events and mortality. Newer potassium binders drugs can prevent / decrease hyperkalemia induced by RAAS blockade, and may reduce the high discontinuation rates or dose reduction of RAAS-blockers. The nonsteroidal MR blockers, with more potency and selectivity than the classic ones, reduce proteinuria and have a lower risk of hyperkalemia. Several clinical trials, currently underway, will determine the effect of classic MR blockers on CV events and mortality in subjects with stage 3b CKD and in dialysis patients, and whether in patients with type 2 diabetes mellitus and CKD, optimally treated and with high risk of CV and kidney events, the addition of finerenone to their treatment produces cardiorenal benefits. Large randomized trials have shown that sodium glucose type 2 cotransporter inhibitors (SGLT2i) reduce mortality and the development and progression of diabetic and nondiabetic CKD. There are pathophysiological arguments, which raise the possibility that the triple combination ACEi or ARBs, SGLT2i and aldosterone antagonist provide additional renal and cardiovascular protection.
Collapse
|
15
|
Brandt-Jacobsen NH, Johansen ML, Rasmussen J, Forman JL, Holm MR, Faber J, Rossignol P, Schou M, Kistorp C. Effect of high-dose mineralocorticoid receptor antagonist eplerenone on urinary albumin excretion in patients with type 2 diabetes and high cardiovascular risk: Data from the MIRAD trial. DIABETES & METABOLISM 2020; 47:101190. [PMID: 32919068 DOI: 10.1016/j.diabet.2020.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 12/20/2022]
Abstract
AIM As mineralocorticoid receptor antagonists (MRAs) may possess renoprotective effects in type 2 diabetes (T2D), it was decided to investigate the impact of high-dose MRA on prespecified secondary endpoints-namely, change in urinary albumin-creatinine ratio (UACR) and 24-h ambulatory blood pressure-in the MIRAD trial. METHODS This was a double-blind clinical trial in which T2D patients at high risk of or with established cardiovascular disease (CVD) were randomized to either high-dose (100-200 mg) eplerenone or a dose-matched placebo as an add-on to background antihypertensive treatment for 26 weeks. Safety was evaluated by the incidence of hyperkalaemia and kidney-related adverse events. RESULTS A total of 140 patients were enrolled (70 in each group). Baseline UACR was 17 mg/g (geometric mean; 95% CI: 13-22); this decreased by 34% in the eplerenone group compared with the placebo group at week 26 (95% CI: -51% to -12%; P = 0.005). There was no significant decrease in 24-h systolic blood pressure (SBP) due to treatment (-3 mmHg; 95% CI: -6 to 1; P = 0.150). However, the observed change in 24-h SBP correlated with the relative change in UACR in the eplerenone group (r = 0.568, P < 0.001). Mean baseline (± SD) estimated glomerular filtration rate (eGFR) was 85 (± 18.6) mL/min/1.73 m2, and 12 (± 9%) had an eGFR of 41-59 mL/min/1.73 m2. No significant differences in the incidence of mild hyperkalaemia (≥ 5.5 mmol/L; eplerenone vs placebo: 6 vs 2, respectively; P = 0.276) and no severe hyperkalaemia (≥ 6.0 mmol/L) were observed. CONCLUSION The addition of high-dose eplerenone to T2D patients at high risk of CVD can markedly reduce UACR with an acceptable safety profile.
Collapse
Affiliation(s)
- Niels H Brandt-Jacobsen
- Department of Endocrinology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Marie Louise Johansen
- Department of Endocrinology-Internal Medicine, Copenhagen University Hospital, Herlev-Gentofte Hospital
| | - Jon Rasmussen
- Department of Endocrinology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Internal Medicine, Holbæk Hospital, Denmark
| | - Julie L Forman
- Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Section of Biostatistics, Department of Public Health, University of Copenhagen, Denmark
| | | | - Jens Faber
- Department of Endocrinology-Internal Medicine, Copenhagen University Hospital, Herlev-Gentofte Hospital; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Patrick Rossignol
- Université de Lorraine, INSERM CIC Plurithémathique 1433, UMRS 1116 INSERM, CHRU Nancy, and FCRIN INI-CRCT, Nancy, France
| | - Morten Schou
- Faculty of Health and Medical Sciences, University of Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital, Herlev-Gentofte Hospital, Denmark
| | - Caroline Kistorp
- Department of Endocrinology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
16
|
Hall JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity, kidney dysfunction and hypertension: mechanistic links. Nat Rev Nephrol 2020; 15:367-385. [PMID: 31015582 DOI: 10.1038/s41581-019-0145-4] [Citation(s) in RCA: 327] [Impact Index Per Article: 65.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Excessive adiposity raises blood pressure and accounts for 65-75% of primary hypertension, which is a major driver of cardiovascular and kidney diseases. In obesity, abnormal kidney function and associated increases in tubular sodium reabsorption initiate hypertension, which is often mild before the development of target organ injury. Factors that contribute to increased sodium reabsorption in obesity include kidney compression by visceral, perirenal and renal sinus fat; increased renal sympathetic nerve activity (RSNA); increased levels of anti-natriuretic hormones, such as angiotensin II and aldosterone; and adipokines, particularly leptin. The renal and neurohormonal pathways of obesity and hypertension are intertwined. For example, leptin increases RSNA by stimulating the central nervous system proopiomelanocortin-melanocortin 4 receptor pathway, and kidney compression and RSNA contribute to renin-angiotensin-aldosterone system activation. Glucocorticoids and/or oxidative stress may also contribute to mineralocorticoid receptor activation in obesity. Prolonged obesity and progressive renal injury often lead to the development of treatment-resistant hypertension. Patient management therefore often requires multiple antihypertensive drugs and concurrent treatment of dyslipidaemia, insulin resistance, diabetes and inflammation. If more effective strategies for the prevention and control of obesity are not developed, cardiorenal, metabolic and other obesity-associated diseases could overwhelm health-care systems in the future.
Collapse
Affiliation(s)
- John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA. .,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
17
|
Chen YY, Lin YHH, Huang WC, Chueh E, Chen L, Yang SY, Lin P, Lin LY, Lin YH, Wu VC, Chu T, Wu KD. Adrenalectomy Improves the Long-Term Risk of End-Stage Renal Disease and Mortality of Primary Aldosteronism. J Endocr Soc 2019; 3:1110-1126. [PMID: 31086833 PMCID: PMC6507624 DOI: 10.1210/js.2019-00019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/19/2019] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Primary aldosteronism (PA) is a common cause of secondary hypertension, and the long-term effect of excess aldosterone on kidney function is unknown. PATIENTS AND METHODS We used a longitudinal population database from the Taiwan National Health Insurance system and applied a validated algorithm to identify patients with PA diagnosed between 1997 and 2009. RESULTS There were 2699 patients with PA recruited, of whom 761 patients with an aldosterone-producing adenoma (APA) were identified. The incidence rate of end-stage renal disease (ESRD) was 3% in patients with PA after targeted treatments and 5.2 years of follow-up, which was comparable to the rate in controls with essential hypertension (EH). However, after taking mortality as a competing risk, we found a significantly lower incidence of ESRD when comparing patients with PA vs EH [subdistribution hazard ratio (sHR), 0.38; P = 0.007] and patients with APA vs EH (sHR 0.55; P = 0.021) after adrenalectomy; however, we did not see similar results in groups with mineralocorticoid receptor antagonist (MRA)‒treated PA vs EH. There was also a significantly lower incidence of mortality in groups with PA and APA who underwent adrenalectomy than among EH controls (P < 0.001). CONCLUSION Regarding incident ESRD, patients with PA were comparable to their EH counterparts after treatment. After adrenalectomy, patients with APA had better long-term outcomes regarding progression to ESRD and mortality than hypertensive controls, but MRA treatments did not significantly affect outcome.
Collapse
Affiliation(s)
- Ying-Ying Chen
- Division of Nephrology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - You-Hsien Hugo Lin
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Wei-Chieh Huang
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, New Taipei City Hospital, New Taipei City, Taiwan
| | - Eric Chueh
- Case Western Reserve University, Cleveland, Ohio
| | - Likwang Chen
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Shao-Yu Yang
- Division of Nephrology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- TAIPAI, Taiwan Primary Aldosteronism Investigator, Taipei, Taiwan
| | - Po‐Chih Lin
- Division of Nephrology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- TAIPAI, Taiwan Primary Aldosteronism Investigator, Taipei, Taiwan
| | - Lian-Yu Lin
- Division of Nephrology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- TAIPAI, Taiwan Primary Aldosteronism Investigator, Taipei, Taiwan
| | - Yen-Hung Lin
- Division of Nephrology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- TAIPAI, Taiwan Primary Aldosteronism Investigator, Taipei, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- TAIPAI, Taiwan Primary Aldosteronism Investigator, Taipei, Taiwan
| | | | - Kwan Dun Wu
- Division of Nephrology and Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- TAIPAI, Taiwan Primary Aldosteronism Investigator, Taipei, Taiwan
| |
Collapse
|
18
|
Zhang J, Wei J, Jiang S, Xu L, Wang L, Cheng F, Buggs J, Koepsell H, Vallon V, Liu R. Macula Densa SGLT1-NOS1-Tubuloglomerular Feedback Pathway, a New Mechanism for Glomerular Hyperfiltration during Hyperglycemia. J Am Soc Nephrol 2019; 30:578-593. [PMID: 30867247 DOI: 10.1681/asn.2018080844] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 01/27/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Glomerular hyperfiltration is common in early diabetes and is considered a risk factor for later diabetic nephropathy. We propose that sodium-glucose cotransporter 1 (SGLT1) senses increases in luminal glucose at the macula densa, enhancing generation of neuronal nitric oxide synthase 1 (NOS1)-dependent nitric oxide (NO) in the macula densa and blunting the tubuloglomerular feedback (TGF) response, thereby promoting the rise in GFR. METHODS We used microperfusion, micropuncture, and renal clearance of FITC-inulin to examine the effects of tubular glucose on NO generation at the macula densa, TGF, and GFR in wild-type and macula densa-specific NOS1 knockout mice. RESULTS Acute intravenous injection of glucose induced hyperglycemia and glucosuria with increased GFR in mice. We found that tubular glucose blunts the TGF response in vivo and in vitro and stimulates NO generation at the macula densa. We also showed that SGLT1 is expressed at the macula densa; in the presence of tubular glucose, SGLT1 inhibits TGF and NO generation, but this action is blocked when the SGLT1 inhibitor KGA-2727 is present. In addition, we demonstrated that glucose increases NOS1 expression and NOS1 phosphorylation at Ser1417 in mouse renal cortex and cultured human kidney tissue. In macula densa-specific NOS1 knockout mice, glucose had no effect on NO generation, TGF, and GFR. CONCLUSIONS We identified a novel mechanism of acute hyperglycemia-induced hyperfiltration wherein increases in luminal glucose at the macula densa upregulate the expression and activity of NOS1 via SGLT1, blunting the TGF response and promoting glomerular hyperfiltration.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Molecular Pharmacology and Physiology, College of Medicine,
| | - Jin Wei
- Department of Molecular Pharmacology and Physiology, College of Medicine
| | - Shan Jiang
- Department of Molecular Pharmacology and Physiology, College of Medicine
| | - Lan Xu
- Department of Biostatistics, College of Public Health, and
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, College of Medicine
| | - Feng Cheng
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, Florida
| | - Jacentha Buggs
- Advanced Organ Disease & Transplantation Institute, Tampa General Hospital, Tampa, Florida
| | - Hermann Koepsell
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany; and
| | - Volker Vallon
- Division of Nephrology and Hypertension, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, College of Medicine
| |
Collapse
|
19
|
Pressly JD, Soni H, Jiang S, Wei J, Liu R, Moore BM, Adebiyi A, Park F. Activation of the cannabinoid receptor 2 increases renal perfusion. Physiol Genomics 2019; 51:90-96. [PMID: 30707046 DOI: 10.1152/physiolgenomics.00001.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Acute kidney injury (AKI) is an increasing clinical problem that is associated with chronic kidney disease progression. Cannabinoid receptor 2 (CB2) activation has been shown to mitigate some of the deleterious tubular effects due to AKI, but its role on the renal vasculature has not been fully described. In this study, we investigated the effects of our novel CB2 receptor agonist, SMM-295, on renal vasculature by assessing cortical perfusion with laser Doppler flowmetry and changes in luminal diameter with isolated afferent arterioles. In this study, intravenously infused SMM-295 (6 mg/kg) significantly increased cortical renal perfusion (13.8 ± 0.6%; P < 0.0001; n = 7) compared with vehicle (0.1 ± 1.5%; n = 10) normalized to baseline values in anesthetized C57BL/6J mice. This effect was not dependent upon activation of the CB1 receptor (met-anandamide; 6 mg/kg iv) and was predominantly abolished in Cnr2 knockout mice with SMM-295 (6 mg/kg iv). Ablation of the renal afferent nerves with capsaicin blocked the SMM-295-dependent increase in renal cortical perfusion, and the increased renal blood flow was not dependent upon products synthesized by cyclooxygenase or nitric oxide synthase. The increased renal perfusion by CB2 receptor activation is also attributed to a direct vascular effect, since SMM-295 (5 μM) engendered a significant 37 ± 7% increase ( P < 0.0001; n = 4) in luminal diameters of norepinephrine-preconstricted afferent arterioles. These data provide new insight into the potential benefit of SMM-295 by activating vascular and nonvascular CB2 receptors to promote renal vasodilation, and provide a new therapeutic target to treat renal injuries that impact renal blood flow dynamics.
Collapse
Affiliation(s)
- J D Pressly
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee
| | - H Soni
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - S Jiang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - J Wei
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - R Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - B M Moore
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee
| | - A Adebiyi
- Department of Physiology, University of Tennessee Health Science Center , Memphis, Tennessee
| | - F Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee
| |
Collapse
|
20
|
Nizar JM, Shepard BD, Vo VT, Bhalla V. Renal tubule insulin receptor modestly promotes elevated blood pressure and markedly stimulates glucose reabsorption. JCI Insight 2018; 3:95107. [PMID: 30135311 DOI: 10.1172/jci.insight.95107] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/03/2018] [Indexed: 12/25/2022] Open
Abstract
Although the cause of hypertension among individuals with obesity and insulin resistance is unknown, increased plasma insulin, acting in the kidney to increase sodium reabsorption, has been proposed as a potential mechanism. Insulin may also stimulate glucose uptake, but the contributions of tubular insulin signaling to sodium or glucose transport in the setting of insulin resistance is unknown. To directly study the role of insulin signaling in the kidney, we generated inducible renal tubule-specific insulin receptor-KO mice and used high-fat feeding and mineralocorticoids to model obesity and insulin resistance. Insulin receptor deletion did not alter blood pressure or sodium excretion in mice on a high-fat diet alone, but it mildly attenuated the increase in blood pressure with mineralocorticoid supplementation. Under these conditions, KO mice developed profound glucosuria. Insulin receptor deletion significantly reduced SGLT2 expression and increased urinary glucose excretion and urine flow. These data demonstrate a direct role for insulin receptor-stimulated sodium and glucose transport and a functional interaction of insulin signaling with mineralocorticoids in vivo. These studies uncover a potential mechanistic link between preserved insulin sensitivity and renal glucose handling in obesity and insulin resistance.
Collapse
Affiliation(s)
- Jonathan M Nizar
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Blythe D Shepard
- Department of Human Science, Georgetown University, Washington, DC
| | - Vianna T Vo
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
21
|
Song J, Wang L, Fan F, Wei J, Zhang J, Lu Y, Fu Y, Wang S, Juncos LA, Liu R. Role of the Primary Cilia on the Macula Densa and Thick Ascending Limbs in Regulation of Sodium Excretion and Hemodynamics. Hypertension 2017; 70:324-333. [PMID: 28607127 PMCID: PMC5507816 DOI: 10.1161/hypertensionaha.117.09584] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 01/28/2023]
Abstract
We investigated the significance of the primary cilia on the macula densa and thick ascending limb (TAL) in regulation of renal hemodynamics, sodium excretion, and blood pressure in this study. A tissue-specific primary cilia knock-out (KO) mouse line was generated by crossing NKCC2-Cre mice with IFT88-Δ/flox mice (NKCC2CRE; IFT88Δ/flox), in which the primary cilia were deleted from the macula densa and TAL. NO generation was measured with a fluorescent dye (4,5-diaminofluorescein diacetate) in isolated perfused juxtaglomerular apparatus. Deletion of the cilia reduced NO production by 56% and 42% in the macula densa and TAL, respectively. NO generation by the macula densa was inhibited by both a nonselective and a selective nitric oxide synthesis inhibitors, whereas TAL-produced NO was inhibited by a nonselective and not by a selective NO synthesis 1 inhibitor. The tubuloglomerular feedback response was enhanced in the KO mice both in vitro measured with isolated perfused juxtaglomerular apparatuses and in vivo measured with micropuncture. In response to an acute volume expansion, the KO mice exhibited limited glomerular filtration rate elevation and impaired sodium excretion compared with the wild-type mice. The mean arterial pressure measured with telemetry was the same for wild-type and KO mice fed a normal salt diet. After a high salt diet, the mean arterial pressure increased by 17.4±1.6 mm Hg in the KO mice. On the basis of these findings, we concluded that the primary cilia on the macula densa and TAL play an essential role in the control of sodium excretion and blood pressure.
Collapse
Affiliation(s)
- Jiangping Song
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Lei Wang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Fan Fan
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Jin Wei
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Jie Zhang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Yan Lu
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Yiling Fu
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Shaohui Wang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Luis A Juncos
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.)
| | - Ruisheng Liu
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (J.S., L.W., J.W., J.Z., S.W., R.L.); State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (J.S.); and Department of Pharmacology and Medicine, University of Mississippi Medical Center, Jackson (F.F., Y.L., Y.F., L.A.J.).
| |
Collapse
|
22
|
Hundemer GL, Baudrand R, Brown JM, Curhan G, Williams GH, Vaidya A. Renin Phenotypes Characterize Vascular Disease, Autonomous Aldosteronism, and Mineralocorticoid Receptor Activity. J Clin Endocrinol Metab 2017; 102:1835-1843. [PMID: 28323995 PMCID: PMC5470762 DOI: 10.1210/jc.2016-3867] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 02/14/2017] [Indexed: 01/13/2023]
Abstract
CONTEXT Mild cases of autonomous aldosterone secretion may go unrecognized using current diagnostic criteria for primary aldosteronism (PA). OBJECTIVE To investigate whether the inability to stimulate renin serves as a biomarker for unrecognized autonomous aldosterone secretion and mineralocorticoid receptor (MR) activation. PARTICIPANTS Six hundred sixty-three normotensive and mildly hypertensive participants, who were confirmed to not have PA using current guideline criteria and were on no antihypertensive medications. DESIGN Participants had their maximally stimulated plasma renin activity (PRA) measured while standing upright after sodium restriction. Tertiles of maximally stimulated PRA were hypothesized to reflect the degree of MR activation: lowest PRA tertile = "Inappropriate/Excess MR Activity;" middle PRA tertile = "Intermediate MR Activity;"; and highest PRA tertile = "Physiologic MR Activity." All participants underwent detailed biochemical and vascular characterizations under conditions of liberalized sodium intake, and associations with stimulated PRA phenotypes were performed. RESULTS Participants with lower stimulated PRA had greater autonomous aldosterone secretion [higher aldosterone-to-renin ratio (P = 0.002), higher urine aldosterone excretion rate (P = 0.003), higher systolic blood pressure (P = 0.004), and lower renal plasma flow (P = 0.04)] and a nonsignificant trend toward lower serum potassium and higher urine potassium excretion, which became significant after stratification by hypertension status. CONCLUSIONS In participants without clinical PA, the inability to stimulate renin was associated with greater autonomous aldosterone secretion, impaired vascular function, and suggestive trends in potassium handling that indicate an extensive spectrum of unrecognized MR activation.
Collapse
Affiliation(s)
- Gregory L. Hundemer
- Division of Renal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Rene Baudrand
- Program for Adrenal Disorders and Endocrine Hypertension, Department of Endocrinology, Pontificia Universidad Catolica de Chile School of Medicine, Santiago, Chile
| | - Jenifer M. Brown
- Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Gary Curhan
- Division of Renal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Gordon H. Williams
- Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| | - Anand Vaidya
- Center for Adrenal Disorders, Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
23
|
Bar-Nur D, Jaber BL. Mineralocorticoid Receptor Blockade for Prevention of Acute Kidney Injury: An Elusive Target. Am J Kidney Dis 2017; 69:166-168. [DOI: 10.1053/j.ajkd.2016.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 11/11/2022]
|
24
|
Wei J, Song J, Jiang S, Zhang G, Wheeler D, Zhang J, Wang S, Lai EY, Wang L, Buggs J, Liu R. Role of intratubular pressure during the ischemic phase in acute kidney injury. Am J Physiol Renal Physiol 2016; 312:F1158-F1165. [PMID: 28579560 DOI: 10.1152/ajprenal.00527.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 10/19/2016] [Accepted: 10/31/2016] [Indexed: 01/25/2023] Open
Abstract
Acute kidney injury (AKI) induced by clamping of renal vein or pedicle is more severe than clamping of artery, but the mechanism has not been clarified. In the present study, we tested our hypothesis that increased proximal tubular pressure (Pt) during the ischemic phase exacerbates kidney injury and promotes the development of AKI. We induced AKI by bilateral clamping of renal arteries, pedicles, or veins for 18 min at 37°C, respectively. Pt during the ischemic phase was measured with micropuncture. We found that higher Pt was associated with more severe AKI. To determine the role of Pt during the ischemic phase on the development of AKI, we adjusted the Pt by altering renal artery pressure. We induced AKI by bilateral clamping of renal veins, and the Pt was changed by adjusting the renal artery pressure during the ischemic phase by constriction of aorta and mesenteric artery. When we decreased renal artery pressure from 85 ± 5 to 65 ± 8 mmHg, Pt decreased from 53.3 ± 2.7 to 44.7 ± 2.0 mmHg. Plasma creatinine decreased from 2.48 ± 0.23 to 1.91 ± 0.21 mg/dl at 24 h after renal ischemia. When we raised renal artery pressure to 103 ± 7 mmHg, Pt increased to 67.2 ± 5.1 mmHg. Plasma creatinine elevated to 3.17 ± 0.14 mg·dl·24 h after renal ischemia. Changes in KIM-1, NGAL, and histology were in the similar pattern as plasma creatinine. In summary, we found that higher Pt during the ischemic phase promoted the development of AKI, while lower Pt protected from kidney injury. Pt may be a potential target for treatment of AKI.
Collapse
Affiliation(s)
- Jin Wei
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Jiangping Song
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida.,State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shan Jiang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida.,Department of Physiology, Zhejiang University School of Medicine, Zhejiang, China
| | - Gensheng Zhang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida.,Department of Physiology, Zhejiang University School of Medicine, Zhejiang, China
| | - Donald Wheeler
- Department of Pathology and Cell Biology, University of South Florida College of Medicine, Tampa, Florida
| | - Jie Zhang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Shaohui Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - En Yin Lai
- Department of Physiology, Zhejiang University School of Medicine, Zhejiang, China
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | | | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida;
| |
Collapse
|
25
|
Ren Y, Janic B, Kutskill K, Peterson EL, Carretero OA. Mechanisms of connecting tubule glomerular feedback enhancement by aldosterone. Am J Physiol Renal Physiol 2016; 311:F1182-F1188. [PMID: 27413197 DOI: 10.1152/ajprenal.00076.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/08/2016] [Indexed: 11/22/2022] Open
Abstract
Connecting tubule glomerular feedback (CTGF) is a mechanism where an increase in sodium (Na) concentration in the connecting tubule (CNT) causes the afferent arteriole (Af-Art) to dilate. We recently reported that aldosterone within the CNT lumen enhances CTGF via a nongenomic effect involving GPR30 receptors and sodium/hydrogen exchanger (NHE), but the signaling pathways of this mechanism are unknown. We hypothesize that aldosterone enhances CTGF via cAMP/protein kinase A (PKA) pathway that activates protein kinase C (PKC) and stimulates superoxide (O2-) production. Rabbit Af-Arts and their adherent CNTs were microdissected and simultaneously perfused. Two consecutive CTGF curves were elicited by increasing the CNT luminal NaCl. We found that the main effect of aldosterone was to sensitize CTGF and we analyzed data by comparing NaCl concentration in the CNT perfusate needed to achieve half of the maximal response (EC50). During the control period, the NaCl concentration that elicited a half-maximal response (EC50) was 37.0 ± 2.0 mmol/l; addition of aldosterone (10-8 mol/l) to the CNT lumen decreased EC50 to 19.3 ± 1.3 mmol/l (P ≤ 0.001 vs. Control). The specific adenylyl cyclase inhibitor 2',3'-dideoxyadenosine (ddA; 2 × 10-4 mol/l) and the PKA inhibitor H-89 dihydrochloride hydrate (H-89; 2 × 10-6 mol/l) prevented the aldosterone effect. The selective PKC inhibitor GF109203X (10-8 mol/l) also prevented EC50 reduction caused by aldosterone. CNT intraluminal addition of O2- scavenger tempol (10-4 mol/l) blocked the aldosterone effect. We conclude that aldosterone inside the CNT lumen enhances CTGF via a cAMP/PKA/PKC pathway and stimulates O2- generation and this process may contribute to renal damage by increasing glomerular capillary pressure.
Collapse
Affiliation(s)
- YiLin Ren
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Branislava Janic
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Kristopher Kutskill
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Edward L Peterson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, Michigan
| | - Oscar A Carretero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| |
Collapse
|
26
|
Wang X, Chandrashekar K, Wang L, Lai EY, Wei J, Zhang G, Wang S, Zhang J, Juncos LA, Liu R. Inhibition of Nitric Oxide Synthase 1 Induces Salt-Sensitive Hypertension in Nitric Oxide Synthase 1α Knockout and Wild-Type Mice. Hypertension 2016; 67:792-9. [PMID: 26883268 DOI: 10.1161/hypertensionaha.115.07032] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/18/2016] [Indexed: 01/10/2023]
Abstract
We recently showed that α, β, and γ splice variants of neuronal nitric oxide synthase (NOS1) expressed in the macula densa and NOS1β accounts for most of the NO generation. We have also demonstrated that the mice with deletion of NOS1 specifically from the macula densa developed salt-sensitive hypertension. However, the global NOS1 knockout (NOS1KO) strain is neither hypertensive nor salt sensitive. This global NOS1KO strain is actually an NOS1αKO model. Consequently, we hypothesized that inhibition of NOS1β in NOS1αKO mice induces salt-sensitive hypertension. NOS1αKO and C57BL/6 wild-type (WT) mice were implanted with telemetry transmitters and divided into 7-nitroindazole (10 mg/kg/d)-treated and nontreated groups. All of the mice were fed a normal salt (0.4% NaCl) diet for 5 days, followed by a high-salt diet (4% NaCl). NO generation by the macula densa was inhibited by >90% in WT and NOS1αKO mice treated with 7-nitroindazole. Glomerular filtration rate in conscious mice was increased by ≈ 40% after a high-salt diet in both NOS1αKO and WT mice. In response to acute volume expansion, glomerular filtration rate, diuretic and natriuretic response were significantly blunted in the WT and knockout mice treated with 7-nitroindazole. Mean arterial pressure had no significant changes in mice fed a high-salt diet, but increased ≈ 15 mm Hg similarly in NOS1αKO and WT mice treated with 7-nitroindazole. We conclude that NOS1β, but not NOS1α, plays an important role in control of sodium excretion and hemodynamics in response to either an acute or a chronic salt loading.
Collapse
Affiliation(s)
- Ximing Wang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Kiran Chandrashekar
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Lei Wang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - En Yin Lai
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Jin Wei
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Gensheng Zhang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Shaohui Wang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Jie Zhang
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Luis A Juncos
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.)
| | - Ruisheng Liu
- From the Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa (X.W., L.W., J.W., G.Z., S.W., J.Z., R.L.); Shandong Medical Imaging Research Institute, Shandong Provincial Key Laboratory of Diagnosis and Treatment of Cardio-Cerebral Vascular Disease, Shandong University, Jinan, Shandong, China (X.W.); Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson (K.C., L.A.J.); and Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China (E.Y.L., G.Z.).
| |
Collapse
|
27
|
Iwakura Y, Ito S, Morimoto R, Kudo M, Ono Y, Nezu M, Takase K, Seiji K, Ishidoya S, Arai Y, Funamizu Y, Miki T, Nakamura Y, Sasano H, Satoh F. Renal Resistive Index Predicts Postoperative Blood Pressure Outcome in Primary Aldosteronism. Hypertension 2016; 67:654-60. [PMID: 26865201 DOI: 10.1161/hypertensionaha.115.05924] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/30/2015] [Indexed: 11/16/2022]
Abstract
The renal resistive index (RI) calculated by Doppler ultrasonography has been reported to be correlated with renal structural changes and outcomes in patients with essential hypertension or renal disease. However, little is known about this index in primary aldosteronism. In this prospective study, we examined the utility of this index to predict blood pressure (BP) outcome after adrenalectomy in patients with primary aldosteronism. We studied 94 patients with histopathologically proven aldosteronoma who underwent surgery. Parameters on renal function, including renal flow indices, were examined and followed up for 12 months postoperatively. The renal RI of the main, hilum, and interlobar arteries was significantly higher in patients with aldosteronoma compared with 100 control patients. BP, estimated glomerular filtration rate, and urinary albumin excretion significantly decreased after adrenalectomy. The resistive indices of all compartment arteries were significantly reduced 1 month after adrenalectomy and remained stable for 12 months. Patients whose interlobar RI was in the highest tertile at baseline had higher systolic BP after adrenalectomy than those whose RI was in the lowest tertile. Logistic regression analysis demonstrated that the RI of the interlobar and hilum arteries could be an independent predictive marker for intractable hypertension (systolic BP ≥140 mm Hg, increased BP, taking ≥3 antihypertensive agents, or increased number of agents) even after adrenalectomy. Therefore, in patients with aldosteronoma, the renal RI indicates partially reversible renal hemodynamics and renal structural damages that would influence postoperative BP outcome.
Collapse
Affiliation(s)
- Yoshitsugu Iwakura
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Sadayoshi Ito
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Ryo Morimoto
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Masataka Kudo
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Yoshikiyo Ono
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Masahiro Nezu
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Kei Takase
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Kazumasa Seiji
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Shigeto Ishidoya
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Yoichi Arai
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Yasuharu Funamizu
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Takashi Miki
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Yasuhiro Nakamura
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Hironobu Sasano
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.)
| | - Fumitoshi Satoh
- From the Endocrine unit of the Division of Nephrology, Endocrinology and Vascular Medicine (Y.I., S.I., R.M., M.K., Y.O., M.N., F.S.), Departments of Diagnostic Radiology (K.T., K.S.) and Urology (S.I, Y.A.), Clinical Physiology Center (Y.F., T.M.), Department of Pathology (Y.N., H.S.), Tohoku University Hospital, Sendai, Japan; and Division of Clinical Hypertension, Endocrinology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Japan (F.S.).
| |
Collapse
|
28
|
Lu Y, Wei J, Stec DE, Roman RJ, Ge Y, Cheng L, Liu EY, Zhang J, Hansen PBL, Fan F, Juncos LA, Wang L, Pollock J, Huang PL, Fu Y, Wang S, Liu R. Macula Densa Nitric Oxide Synthase 1β Protects against Salt-Sensitive Hypertension. J Am Soc Nephrol 2015; 27:2346-56. [PMID: 26647426 DOI: 10.1681/asn.2015050515] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/24/2015] [Indexed: 01/10/2023] Open
Abstract
Nitric oxide (NO) is an important negative modulator of tubuloglomerular feedback responsiveness. We recently found that macula densa expresses α-, β-, and γ-splice variants of neuronal nitric oxide synthase 1 (NOS1), and NOS1β expression in the macula densa increases on a high-salt diet. This study tested whether upregulation of NOS1β expression in the macula densa affects sodium excretion and salt-sensitive hypertension by decreasing tubuloglomerular feedback responsiveness. Expression levels of NOS1β mRNA and protein were 30- and five-fold higher, respectively, than those of NOS1α in the renal cortex of C57BL/6 mice. Furthermore, macula densa NO production was similar in the isolated perfused juxtaglomerular apparatus of wild-type (WT) and nitric oxide synthase 1α-knockout (NOS1αKO) mice. Compared with control mice, mice with macula densa-specific knockout of all nitric oxide synthase 1 isoforms (MD-NOS1KO) had a significantly enhanced tubuloglomerular feedback response and after acute volume expansion, significantly reduced GFR, urine flow, and sodium excretion. Mean arterial pressure increased significantly in MD-NOS1KO mice (P<0.01) but not NOS1flox/flox mice fed a high-salt diet. After infusion of angiotensin II, mean arterial pressure increased by 61.6 mmHg in MD-NOS1KO mice versus 32.0 mmHg in WT mice (P<0.01) fed a high-salt diet. These results indicate that NOS1β is a primary NOS1 isoform expressed in the macula densa and regulates the tubuloglomerular feedback response, the natriuretic response to acute volume expansion, and the development of salt-sensitive hypertension. These findings show a novel mechanism for salt sensitivity of BP and the significance of tubuloglomerular feedback response in long-term control of sodium excretion and BP.
Collapse
Affiliation(s)
- Yan Lu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida; Departments of Physiology and Biophysics and
| | - Jin Wei
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | | | - Richard J Roman
- Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ying Ge
- Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Liang Cheng
- Departments of Physiology and Biophysics and
| | - Eddie Y Liu
- Departments of Physiology and Biophysics and
| | - Jie Zhang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | | | - Fan Fan
- Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | | | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Jennifer Pollock
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Paul L Huang
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yiling Fu
- Departments of Physiology and Biophysics and
| | - Shaohui Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine, Tampa, Florida; Departments of Physiology and Biophysics and
| |
Collapse
|
29
|
Layton AT. Recent advances in renal hemodynamics: insights from bench experiments and computer simulations. Am J Physiol Renal Physiol 2015; 308:F951-5. [PMID: 25715984 DOI: 10.1152/ajprenal.00008.2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/23/2015] [Indexed: 01/08/2023] Open
Abstract
It has been long known that the kidney plays an essential role in the control of body fluids and blood pressure and that impairment of renal function may lead to the development of diseases such as hypertension (Guyton AC, Coleman TG, Granger Annu Rev Physiol 34: 13-46, 1972). In this review, we highlight recent advances in our understanding of renal hemodynamics, obtained from experimental and theoretical studies. Some of these studies were published in response to a recent Call for Papers of this journal: Renal Hemodynamics: Integrating with the Nephron and Beyond.
Collapse
Affiliation(s)
- Anita T Layton
- Department of Mathematics, Duke University, Durham, North Carolina
| |
Collapse
|
30
|
Liao MT, Wu XM, Chang CC, Liao CW, Chen YH, Lu CC, Lin YT, Chang YY, Hung CS, Lin LC, Lai CL, Lin LY, Wu VC, Ho YL, Wu KD, Lin YH. The Association between Glomerular Hyperfiltration and Left Ventricular Structure and Function in Patients with Primary Aldosteronism. Int J Med Sci 2015; 12:369-77. [PMID: 26005371 PMCID: PMC4441061 DOI: 10.7150/ijms.10975] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 04/20/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Glomerular hyperfiltration has been recently noticed as an important issue in primary aldosteronism (PA) patients. However, its effect on the cardiovascular system remains unknown. METHODS We prospectively analyzed 47 PA patients including 11 PA patients with estimated glomerular filtration rate (eGFR) > 130 ml/min per 1.73 m2 (group 1), and 36 PA patients with eGFR 90-110 ml/min per 1.73 m2 (group 2). Fourteen essential hypertension (EH) patients with eGFR 90-110 ml/min per 1.73 m2 were included as the control group (group 3). Echocardiography including left ventricular mass index (LVMI) measurement and tissue Doppler imaging (TDI) was performed. Predicted left ventricular mass (LVM) was calculated. Inappropriate LVM was defined as an excess of > 35% from the predicted value. RESULTS The value of LVMI decreased significantly in order from groups 1 to 3 (group 1>2>3). While group 2 had a significantly higher percentage of inappropriate LVM than group 3, the percentage of inappropriate LVM were comparable in groups 1 and 2. Group 1 had a higher mitral E velocity, E/A ratio than that of group 2. In the TDI study, the E/E' ratio also decreased significantly in order from groups 1 to 3 (group 1>2>3). Group 2 had lower E' than that of group 3, although the E' of group 1 and 2 were comparable. CONCLUSIONS Although PA patients with glomerular hyperfiltration were associated with higher LVMI, higher mitral E velocity, higher E/E' ratio, they had comparable E' with PA patients with normal GFR. This phenomenon may be explained by higher intravascular volume in this patient group.
Collapse
Affiliation(s)
- Min-Tsun Liao
- 1. Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Xue-Ming Wu
- 2. Department of Internal Medicine, Taoyuan General Hospital, Taoyuan, Taiwan
| | - Chin-Chen Chang
- 3. Department of Medical Image, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Che-Wei Liao
- 1. Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Ying-Hsien Chen
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Ching-Chu Lu
- 5. Department of Nuclear Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Yen-Ting Lin
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Yi-Yao Chang
- 6. Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Chi-Sheng Hung
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Lung-Chun Lin
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Chao-Lun Lai
- 1. Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Lian-Yu Lin
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Vin-Cent Wu
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Yi-Lwun Ho
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Kwan-Dun Wu
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | - Yen-Hung Lin
- 4. Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine. Taipei, Taiwan
| | | |
Collapse
|
31
|
Song J, Lu Y, Lai EY, Wei J, Wang L, Chandrashekar K, Wang S, Shen C, Juncos LA, Liu R. Oxidative status in the macula densa modulates tubuloglomerular feedback responsiveness in angiotensin II-induced hypertension. Acta Physiol (Oxf) 2015; 213:249-58. [PMID: 25089004 DOI: 10.1111/apha.12358] [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] [Received: 06/27/2014] [Revised: 06/27/2014] [Accepted: 07/28/2014] [Indexed: 12/20/2022]
Abstract
AIM Tubuloglomerular feedback (TGF) is an important mechanism in control of signal nephron glomerular filtration rate. The oxidative stress in the macula densa, primarily determined by the interactions between nitric oxide (NO) and superoxide (O2-), is essential in maintaining the TGF responsiveness. However, few studies examining the interactions between and amount of NO and O2- generated by the macula densa during normal and hypertensive states. METHODS In this study, we used isolated perfused juxtaglomerular apparatus to directly measure the amount and also studied the interactions between NO and O2- in macula densa in both physiological and slow pressor Angiotensin II (Ang II)-induced hypertensive mice. RESULTS We found that slow pressor Ang II at a dose of 600 ng kg(-1) min(-1) for two weeks increased mean arterial pressure by 26.1 ± 5.7 mmHg. TGF response increased from 3.4 ± 0.2 μm in control to 5.2 ± 0.2 μm in hypertensive mice. We first measured O2- generation by the macula densa and found it was undetectable in control mice. However, O2- generation by the macula densa increased to 21.4 ± 2.5 unit min(-1) in Ang II-induced hypertensive mice. We then measured NO generation and found that NO generation by the macula densa was 138.5 ± 9.3 unit min(-1) in control mice. The NO was undetectable in the macula densa in hypertensive mice infused with Ang II. CONCLUSIONS Under physiological conditions, TGF response is mainly controlled by the NO generated in the macula densa; in Ang II induced hypertension, the TGF response is mainly controlled by the O2- generated by the macula densa.
Collapse
Affiliation(s)
- J. Song
- State Key Laboratory of Cardiovascular Disease; Fuwai Hospital; National Center for Cardiovascular Diseases; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
| | - Y. Lu
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
- Division of Nephrology; Department of Medicine; University of Mississippi Medical Center; Jackson MS USA
| | - E. Y. Lai
- Department of Physiology; Zhejiang University; Hanzhou China
| | - J. Wei
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
| | - L. Wang
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
| | - K. Chandrashekar
- Division of Nephrology; Department of Medicine; University of Mississippi Medical Center; Jackson MS USA
| | - S. Wang
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
| | - C. Shen
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
| | - L. A. Juncos
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
- Division of Nephrology; Department of Medicine; University of Mississippi Medical Center; Jackson MS USA
| | - R. Liu
- Department of Physiology & Biophysics; University of Mississippi Medical Center; Jackson MS USA
- Division of Nephrology; Department of Medicine; University of Mississippi Medical Center; Jackson MS USA
| |
Collapse
|
32
|
Ando K, Ohtsu H, Uchida S, Kaname S, Arakawa Y, Fujita T. Anti-albuminuric effect of the aldosterone blocker eplerenone in non-diabetic hypertensive patients with albuminuria: a double-blind, randomised, placebo-controlled trial. Lancet Diabetes Endocrinol 2014; 2:944-53. [PMID: 25466242 DOI: 10.1016/s2213-8587(14)70194-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND Renin-angiotensin system inhibitors have renoprotective effects in patients with chronic kidney disease, but most patients treated with these drugs have residual urinary albumin excretion. Some small clinical studies show that mineralocorticoid receptor blockade reduces albuminuria. Our study aimed to examine the beneficial effects of addition of a selective aldosterone antagonist, eplerenone, to renin-angiotensin system inhibitors in hypertensive patients with non-diabetic chronic kidney disease. METHODS In this double-blind, randomised, placebo-controlled trial, we enrolled hypertensive patients, aged 20–79 years, with albuminuria (urinary albumin-to-creatinine ratio [UACR] in the first morning void urine of 30–599 mg/g), an estimated glomerular filtration rate of 50 mL/min per 1·73 m2 or more, and who had received an angiotensin-converting enzyme inhibitor, an angiotensin receptor blocker, or both, for at least 8 weeks. Participants were from 59 clinics and hospitals in Japan. Eligible patients were randomly assigned (1:1), stratified by baseline characteristics, to either low-dose eplerenone (50 mg/day) or placebo, with continuation of standard antihypertensive treatment to attain therapeutic goals (<130/80 mm Hg) for 52 weeks. We assessed efficacy in all patients who received allocated treatment, provided a baseline and post-treatment urine sample, and remained in follow-up. We assessed safety in all patients who received allocated treatment. The primary efficacy measure was percent change in UACR in the first morning void urine at week 52 from baseline. The trial is registered at the clinical trials registry of University Hospital Medical Information Network (UMIN), trial identification number UMIN000001803. FINDINGS Between April 1, 2009, and March 31, 2012, we randomly allocated 170 patients to the eplerenone group and 166 patients to the placebo group. In the primary efficacy analysis, mean percent change in UACR from baseline was −17·3% (95% CI −33·65 to −0·94) for 158 patients in the eplerenone group compared with 10·3% (−6·75 to 22·3) for 146 patients in the placebo group (absolute difference −27·6% [–51·15 to −3·96]; p=0·0222). In the safety analyses, 53 (31%) of 169 patients in the eplerenone group had adverse events (five serious), as did 49 (30%) of 163 in the placebo group (seven serious). Although mean serum potassium concentration was higher in the eplerenone group than the placebo group, severe hyperkalaemia (>5·5 mmol/L) was not recorded in either group. INTERPRETATION Addition of low-dose eplerenone to renin-angiotensin system inhibitors might have renoprotective effects through reduction of albuminuria in hypertensive patients with non-diabetic chronic kidney disease, without serious safety concerns. FUNDING Pfizer.
Collapse
|
33
|
Ren Y, D'Ambrosio MA, Garvin JL, Leung P, Kutskill K, Wang H, Peterson EL, Carretero OA. Aldosterone sensitizes connecting tubule glomerular feedback via the aldosterone receptor GPR30. Am J Physiol Renal Physiol 2014; 307:F427-34. [PMID: 24966088 DOI: 10.1152/ajprenal.00072.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Increasing Na delivery to epithelial Na channels (ENaC) in the connecting tubule (CNT) dilates the afferent arteriole (Af-Art), a process we call connecting tubule glomerular feedback (CTGF). We hypothesize that aldosterone sensitizes CTGF via a nongenomic mechanism that stimulates CNT ENaC via the aldosterone receptor GPR30. Rabbit Af-Arts and their adherent CNTs were microdissected and simultaneously perfused. Two consecutive CTGF curves were elicited by increasing luminal NaCl in the CNT. During the control period, the concentration of NaCl that elicited a half-maximal response (EC50) was 37.0 ± 2.0 mmol/l; addition of aldosterone 10(-8) mol/l to the CNT lumen caused a left-shift (decrease) in EC50 to 19.3 ± 1.3 mmol/l (P = 0.001 vs. control; n = 6). Neither the transcription inhibitor actinomycin D nor the translation inhibitor cycloheximide prevented the effect of aldosterone (control EC50 = 34.7 ± 1.9 mmol/l; aldosterone+actinomycin D EC50 = 22.6 ± 1.6 mmol/l; P < 0.001 and control EC50 = 32.4 ± 4.3 mmol/l; aldosterone+cycloheximide EC50 = 17.4 ± 3.3 mmol/l; P < 0.001). The aldosterone antagonist eplerenone prevented the sensitization of CTGF by aldosterone (control EC50 = 33.2 ± 1.7 mmol/l; aldosterone+eplerenone EC50 = 33.5 ± 1.3 mmol/l; n = 7). The GPR30 receptor blocker G-36 blocked the sensitization of CTGF by aldosterone (aldosterone EC50 = 16.5 ± 1.9 mmol/l; aldosterone+G-36 EC50 = 29.0 ± 2.1 mmol/l; n = 7; P < 0.001). Finally, we found that the sensitization of CTGF by aldosterone was mediated, at least in part, by the sodium/hydrogen exchanger (NHE). We conclude that aldosterone in the CNT lumen sensitizes CTGF via a nongenomic effect involving GPR30 receptors and NHE. Sensitized CTGF induced by aldosterone may contribute to renal damage by increasing Af-Art dilation and glomerular capillary pressure (glomerular barotrauma).
Collapse
Affiliation(s)
- YiLin Ren
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Martin A D'Ambrosio
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Jeffrey L Garvin
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio; and
| | - Pablo Leung
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Kristopher Kutskill
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Hong Wang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Edward L Peterson
- Department of Public Health Sciences, Henry Ford Hospital, Detroit, Michigan
| | - Oscar A Carretero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan;
| |
Collapse
|
34
|
Jönsson S, Agic MB, Narfström F, Melville JM, Hultström M. Renal neurohormonal regulation in heart failure decompensation. Am J Physiol Regul Integr Comp Physiol 2014; 307:R493-7. [PMID: 24920735 DOI: 10.1152/ajpregu.00178.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Decompensation in heart failure occurs when the heart fails to balance venous return with cardiac output, leading to fluid congestion and contributing to mortality. Decompensated heart failure can cause acute kidney injury (AKI), which further increases mortality. Heart failure activates signaling systems that are deleterious to kidneys such as renal sympathetic nerve activity (RSNA), renin-angiotensin-aldosterone system, and vasopressin secretion. All three reduce renal blood flow (RBF) and increase tubular sodium reabsorption, which may increase renal oxygen consumption causing AKI through renal tissue hypoxia. Vasopressin contributes to venous congestion through aquaporin-mediated water retention. Additional water retention may be mediated through vasopressin-induced medullary urea transport and hyaluronan but needs further study. In addition, there are several systems that could protect the kidneys and reduce fluid retention such as natriuretic peptides, prostaglandins, and nitric oxide. However, the effect of natriuretic peptides and nitric oxide are blunted in decompensation, partly due to oxidative stress. This review considers how neurohormonal signaling in heart failure drives fluid retention by the kidneys and thus exacerbates decompensation. It further identifies areas where there is limited data, such as signaling systems 20-HETE, purines, endothelin, the role of renal water retention mechanisms for congestion, and renal hypoxia in AKI during heart failure.
Collapse
Affiliation(s)
- Sofia Jönsson
- Unit for Integrative Physiology, Department of Medical Cellbiology, Uppsala University, Uppsala, Sweden; and
| | - Mediha Becirovic Agic
- Unit for Integrative Physiology, Department of Medical Cellbiology, Uppsala University, Uppsala, Sweden; and
| | - Fredrik Narfström
- Unit for Anaesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jacqueline M Melville
- Unit for Integrative Physiology, Department of Medical Cellbiology, Uppsala University, Uppsala, Sweden; and
| | - Michael Hultström
- Unit for Integrative Physiology, Department of Medical Cellbiology, Uppsala University, Uppsala, Sweden; and Unit for Anaesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
35
|
Liu ZZ, Schmerbach K, Lu Y, Perlewitz A, Nikitina T, Cantow K, Seeliger E, Persson PB, Patzak A, Liu R, Sendeski MM. Iodinated contrast media cause direct tubular cell damage, leading to oxidative stress, low nitric oxide, and impairment of tubuloglomerular feedback. Am J Physiol Renal Physiol 2014; 306:F864-72. [PMID: 24431205 DOI: 10.1152/ajprenal.00302.2013] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Iodinated contrast media (CM) have adverse effects that may result in contrast-induced acute kidney injury. Oxidative stress is believed to play a role in CM-induced kidney injury. We test the hypothesis that oxidative stress and reduced nitric oxide in tubules are consequences of CM-induced direct cell damage and that increased local oxidative stress may increase tubuloglomerular feedback. Rat thick ascending limbs (TAL) were isolated and perfused. Superoxide and nitric oxide were quantified using fluorescence techniques. Cell death rate was estimated using propidium iodide and trypan blue. The function of macula densa and tubuloglomerular feedback responsiveness were measured in isolated, perfused juxtaglomerular apparatuses (JGA) of rabbits. The expression of genes related to oxidative stress and the activity of superoxide dismutase (SOD) were investigated in the renal medulla of rats that received CM. CM increased superoxide concentration and reduced nitric oxide bioavailability in TAL. Propidium iodide fluorescence and trypan blue uptake increased more in CM-perfused TAL than in controls, indicating increased rate of cell death. There were no marked acute changes in the expression of genes related to oxidative stress in medullary segments of Henle's loop. SOD activity did not differ between CM and control groups. The tubuloglomerular feedback in isolated JGA was increased by CM. Tubular cell damage and accompanying oxidative stress in our model are consequences of CM-induced direct cell damage, which also modifies the tubulovascular interaction at the macula densa, and may therefore contribute to disturbances of renal perfusion and filtration.
Collapse
Affiliation(s)
- Zhi Zhao Liu
- Institut für Vegetative Physiologie, Charité, Universitätsmedizin Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhang J, Chandrashekar K, Lu Y, Duan Y, Qu P, Wei J, Juncos LA, Liu R. Enhanced expression and activity of Nox2 and Nox4 in the macula densa in ANG II-induced hypertensive mice. Am J Physiol Renal Physiol 2013; 306:F344-50. [PMID: 24285500 DOI: 10.1152/ajprenal.00515.2013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
NAD(P)H oxidase (Nox)2 and Nox4 are the isoforms of Nox expressed in the macula densa (MD). MD-derived superoxide (O₂⁻), primarily generated by Nox2, is enhanced by acute ANG II stimulation. However, the effects of chronic elevations in ANG II during ANG II-induced hypertension on MD-derived O₂⁻ are unknown. We infused a slow pressor dose of ANG II (600 ng·min⁻¹·kg⁻¹) for 2 wk in C57BL/6 mice and found that mean arterial pressure was elevated by 22.3 ± 3.4 mmHg (P < 0.01). We measured O₂⁻ generation in isolated and perfused MDs and found that O₂⁻ generation by the MD was increased from 9.4 ± 0.9 U/min in control mice to 34.7 ± 1.8 U/min in ANG II-induced hypertensive mice (P < 0.01). We stimulated MMDD1 cells, a MD-like cell line, with ANG II and found that O₂⁻ generation increased from 921 ± 91 to 3,687 ± 183 U·min⁻¹·10⁵ cells⁻¹, which was inhibited with apocynin, oxypurinol, or NS-398 by 46%, 14%, and 12%, respectively. We isolated MD cells using laser capture microdissection and measured mRNA levels of Nox. Nox2 and Nox4 levels increased by 3.7 ± 0.17- and 2.6 ± 0.15-fold in ANG II-infused mice compared with control mice. In MMDD1 cells treated with Nox2 or Nox4 small interfering (si)RNAs, ANG II-stimulated O₂⁻ generation was blunted by 50% and 41%, respectively. In cells treated with p22(phox) siRNA, ANG II-stimulated O₂⁻ generation was completely blocked. In conclusion, we found that a subpressor dose of ANG II enhances O₂⁻ generation in the MD and that the sources of this O₂⁻ are primarily Nox2 and Nox4.
Collapse
Affiliation(s)
- Jie Zhang
- Dept. of Physiology and Biophysics, Univ. of Mississippi Medical Center, 2500 N. State St., Jackson, MS 39216.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Ritz E, Tomaschitz A. Aldosterone and the kidney: a rapidly moving frontier (an update). Nephrol Dial Transplant 2013; 29:2012-9. [PMID: 24194611 DOI: 10.1093/ndt/gft035] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Beyond the classical effect of aldosterone on sodium reabsorption in the distal nephron, the spectrum of aldosterone-induced effects on the kidney (and the cardiovascular system) continues to expand at a rapid pace. Blockade of this system has become an attractive target for intervention. Major contributions have been reported in the past 2-3 years. By necessity this brief summary addresses only some of the emerging issues of nephrological relevance. In this fast moving field, we try to give a concise discussion of papers with potential nephrological relevance in the past 2-3 years.
Collapse
Affiliation(s)
- Eberhard Ritz
- Nierenzentrum, Im Neuenheimer Feld 162, Heidelberg, Germany
| | - Andreas Tomaschitz
- Department of Cardiology, Medical University Graz, Graz, Austria Specialist Clinic for Rehabilitation PV Bad Aussee, Bad Aussee, Austria
| |
Collapse
|
38
|
Brewer J, Liu R, Lu Y, Scott J, Wallace K, Wallukat G, Moseley J, Herse F, Dechend R, Martin JN, Lamarca B. Endothelin-1, oxidative stress, and endogenous angiotensin II: mechanisms of angiotensin II type I receptor autoantibody-enhanced renal and blood pressure response during pregnancy. Hypertension 2013; 62:886-92. [PMID: 24041954 DOI: 10.1161/hypertensionaha.113.01648] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hypertension during preeclampsia is associated with increased maternal vascular sensitivity to angiotensin II (ANGII). This study was designed to determine mechanisms whereby agonistic autoantibodies to the ANGII type I receptor (AT1-AA) enhance blood pressure (mean arterial pressure [MAP]) and renal vascular sensitivity to ANGII during pregnancy. First, we examined MAP and renal artery resistance index in response to chronic administration of ANGII or AT1-AA or AT1-AA+ANGII in pregnant rats compared with control pregnant rats. To examine mechanisms of heightened sensitivity in response to AT1-AA during pregnancy, we examined the role of endogenous ANGII in AT1-AA-infused pregnant rats, and that of endothelin-1 and oxidative stress in AT1-AA+ANGII-treated rats. Chronic ANGII increased MAP from 95±2 in normal pregnant rats to 115±2 mm Hg; chronic AT1-AA increased MAP to 118±1 mm Hg in normal pregnant rats, which further increased to 123±2 mm Hg with AT1-AA+ANGII. Increasing ANGII from 10(-11) to 10(-8) decreased afferent arteriole diameter from 15% to 20% but sharply decreased afferent arteriole diameter to 60% in AT1-AA-pretreated vessels. Renal artery resistance index increased from 0.67 in normal pregnant rats to 0.70 with AT1-AA infusion, which was exacerbated to 0.74 in AT1-AA+ANGII-infused rats. AT1-AA-induced hypertension decreased with enalapril but was not attenuated. Both tissue endothelin-1 and reactive oxygen species increased with AT1-AA+ANGII compared with AT1-AA alone, and blockade of either of these pathways had significant effects on MAP or renal artery resistance index. These data support the hypothesis that AT1-AA, via activation of endothelin-1 and oxidative stress and interaction with endogenous ANGII, is an important mechanism whereby MAP and renal vascular responses are enhanced during preeclampsia.
Collapse
Affiliation(s)
- Justin Brewer
- Department of Pharmacology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hultström M. Neurohormonal interactions on the renal oxygen delivery and consumption in haemorrhagic shock-induced acute kidney injury. Acta Physiol (Oxf) 2013; 209:11-25. [PMID: 23837642 DOI: 10.1111/apha.12147] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/02/2013] [Accepted: 07/04/2013] [Indexed: 12/14/2022]
Abstract
Haemorrhagic shock is a common cause of acute kidney injury (AKI), which is a major risk factor for developing chronic kidney disease. The mechanism is superficially straightforward. An arterial pressure below the kidney's autoregulatory region leads to a direct reduction in filtration pressure and perfusion, which in turn cause renal failure with reduced glomerular filtration rate and AKI because of hypoxia. However, the kidney's situation is further worsened by the hormonal and neural reactions to reduced perfusion pressure. There are three major systems working to maintain arterial pressure in shock: sympathetic signalling, the renin-angiotensin system and vasopressin. These work to retain electrolytes and water and to increase peripheral resistance and cardiac output. In the kidney, the increased electrolyte reabsorption consumes oxygen. At the same time, at the signalling level seen in shock, all of these hormones reduce renal perfusion and thereby oxygen delivery. This creates an exaggerated hypoxic situation that is liable to worsen the AKI. The present review will examine this mechanistic background and identify a number of areas that require further studies. At this time, the ideal treatment of haemorrhagic shock appears to be slow fluid resuscitation, possibly with hyperosmolar sodium, low chloride and no artificial colloids. From the standpoint of the kidney, renin-angiotensin system inhibitors appear fruitful for further study.
Collapse
Affiliation(s)
- M Hultström
- Unit for Integrative Physiology, Department of Medical Cellbiology, Uppsala University, Uppsala, Sweden; Anaesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW The aldosterone/mineralocorticoid receptor system plays an important role in the long-term blood pressure control through Na homeostasis. Its overactivation has been implicated in salt-sensitive hypertension. Excessive salt intake augments the function of mineralocorticoid receptor, despite lowering circulating aldosterone levels, but the mechanism had long been elusive. Recently, Rac1, a member of Rho family small GTP-binding proteins, has emerged as a novel ligand-independent modulator of mineralocorticoid receptor activity. In this review, the roles of Rac1 in the pathogenesis of salt-sensitive hypertension and kidney injury have been summarized. RECENT FINDINGS Genetic engineering studies have highlighted the new aspects of Rac1 and its regulators in salt-sensitive hypertension and cardiac and renal disease. New evidence shows the essential roles of Rac1 in salt-evoked paradoxical mineralocorticoid receptor activation observed in salt-sensitive models and in renal tubular Na reabsorption through reduced nicotinamide-adenine dinucleotide phosphate oxidase-mediated oxidative stress or direct regulation of Na transporters. SUMMARY The emerging concept of 'ligand-independent aberrant mineralocorticoid receptor activation by Rac1' in the pathogenesis of salt-sensitive hypertension and kidney injury has been reviewed. Rac inhibition, in addition to mineralocorticoid receptor blockade and salt restriction, would be a new promising strategy for the treatment of salt-sensitive hypertension.
Collapse
|
41
|
Fu Y, Lu Y, Liu EY, Zhu X, Mahajan GJ, Lu D, Roman RJ, Liu R. Testosterone enhances tubuloglomerular feedback by increasing superoxide production in the macula densa. Am J Physiol Regul Integr Comp Physiol 2013; 304:R726-33. [PMID: 23467324 DOI: 10.1152/ajpregu.00341.2012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Males have higher prevalence of hypertension and renal injury than females, which may be attributed in part to androgen-mediated effects on renal hemodynamics. Tubuloglomerular feedback (TGF) is an important mechanism in control of renal microcirculation. The present study examines the role of testosterone in the regulation of TGF responses. TGF was measured by micropuncture (change of stop-flow pressure, ΔPsf) in castrated Sprague-Dawley rats. The addition of testosterone (10(-7) mol/l) into the lumen increased the ΔPsf from 10.1 ± 1.2 to 12.2 ± 1.2 mmHg. To determine whether androgen receptors (AR) are involved, mRNA of AR was measured in the macula dense cells isolated by laser capture microdissection from kidneys, and a macula densa-like cell line (MMDD1). AR mRNA was expressed in the macula densa of rats and in MMDD1 cells. We next examined the effects of the AR blocker, flutamide (10(-5) mol/l) on the TGF response. The addition of flutamide blocked the effects of testosterone on TGF. The addition of Tempol (10(-4) mol/l) or polyethylene glycol-superoxide dismutase (100 U/ml) to scavenge superoxide blocked the effect of testosterone to augment TGF. We then applied apocynin to inhibit NAD(P)H oxidase and oxypurinol to inhibit xanthine oxidase and found the testosterone-induced augmentation of TGF was blocked. In additional experiments in MMDD1 cells, we found that testosterone increased O2(-) generation. Apocynin or oxypurinol blocked the testosterone-induced increases of O2(-), while blockade of COX-2 with NS-398 had no effect. These findings suggest that testosterone enhances TGF response by stimulating O2(-) production in macula densa via an AR-dependent pathway.
Collapse
Affiliation(s)
- Yiling Fu
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhang Q, Lin L, Lu Y, Liu H, Duan Y, Zhu X, Zou C, Manning RD, Liu R. Interaction between nitric oxide and superoxide in the macula densa in aldosterone-induced alterations of tubuloglomerular feedback. Am J Physiol Renal Physiol 2012; 304:F326-32. [PMID: 23220724 DOI: 10.1152/ajprenal.00501.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Tubuloglomerular feedback (TGF)-mediated constriction of the afferent arteriole is modulated by a balance between release of superoxide (O(2)(-)) and nitric oxide (NO) in macula densa (MD) cells. Aldosterone activates mineralocorticoid receptors that are expressed in the MD and induces both NO and O(2)(-) generation. We hypothesize that aldosterone enhances O(2)(-) production in the MD mediated by protein kinase C (PKC), which buffers the effect of NO in control of TGF response. Studies were performed in microdissected and perfused MD and in a MD cell line, MMDD1 cells. Aldosterone significantly enhanced O(2)(-) generation both in perfused MD and in MMDD1 cells. When aldosterone (10(-7) mol/l) was added in the tubular perfusate, TGF response was reduced from 2.4 ± 0.3 μm to 1.4 ± 0.2 μm in isolated perfused MD. In the presence of tempol, a O(2)(-) scavenger, TGF response was 1.5 ± 0.2 μm. In the presence of both tempol and aldosterone in the tubular perfusate, TGF response was further reduced to 0.4 ± 0.2 μm. To determine if PKC is involved in aldosterone-induced O(2)(-) production, we exposed the O(2)(-) cells to a nonselective PKC inhibitor chelerythrine chloride, a specific PKCα inhibitor Go6976, or a PKCα siRNA, and the aldosterone-induced increase in O(2)(-) production was blocked. These data indicate that aldosterone-stimulated O(2)(-) production in the MD buffers the effect of NO in control of TGF response, an effect that was mediated by PKCα.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kikkawa K. [Revisiting the therapeutic concept of a mineralocorticoid receptor antagonist - focusing on resistant hypertension and chronic renal failure]. Nihon Yakurigaku Zasshi 2012; 139:241-245. [PMID: 22728985 DOI: 10.1254/fpj.139.241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
|