1
|
Flores J, Nugent K. Sodium, the Vascular Endothelium, and Hypertension: A Narrative Review of Literature. Cardiol Rev 2025:00045415-990000000-00402. [PMID: 39807866 DOI: 10.1097/crd.0000000000000854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The vascular endothelium and its endothelial glycocalyx contribute to the protection of the endothelial cells from exposure to high levels of sodium and help these structures maintain normal function by regulating vascular permeability due to its buffering effect. The endothelial glycocalyx has negative surface charges that bind sodium and limit sodium entry into cells and the interstitial space. High sodium levels can disrupt this barrier and allow the movement of sodium into cells and extravascular fluid. This can generate reactive oxygen species that inhibit nitric oxide production. This leads to vasospasm and increases intravascular pressures. Overtime vascular remodeling occurs, and this changes the anatomy of blood vessels, their intrinsic stiffness, and their response to vasodilators and results in hypertension. Patients with increased salt sensitivity are potentially at more risk for this sequence of events. Studies on the degradation of the glycocalyx provide insight into the pathogenesis of clinical disorders with vascular involvement, but there is limited information available in the context of higher concentrations of sodium. Data on higher intake of sodium and the imbalance between nitric oxide and reactive oxygen species have been obtained in experimental studies and provide insights into possible outcomes in humans. The current western diet with sodium intake above recommended levels has led to the assessment of sodium sensitivity, which has been used in different populations and could become a practical tool to evaluate patients. This would potentially allow more focused recommendations regarding salt intake. This review will consider the structure of the vascular endothelium, its components, the effect of sodium on it, and the use of the salt blood test mini.
Collapse
Affiliation(s)
- Jackeline Flores
- From the Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | | |
Collapse
|
2
|
He L, Zuo Q, Ma S, Zhang G, Wang Z, Zhang T, Zhai J, Guo Y. Canagliflozin attenuates kidney injury, gut-derived toxins, and gut microbiota imbalance in high-salt diet-fed Dahl salt-sensitive rats. Ren Fail 2024; 46:2300314. [PMID: 38189082 PMCID: PMC10776083 DOI: 10.1080/0886022x.2023.2300314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/24/2023] [Indexed: 01/09/2024] Open
Abstract
PURPOSE To investigate the effects of canagliflozin (20 mg/kg) on Dahl salt-sensitive (DSS) rat gut microbiota and salt-sensitive hypertension-induced kidney injury and further explore its possible mechanism. METHODS Rats were fed a high-salt diet to induce hypertension and kidney injury, and physical and physiological indicators were measured afterwards. This study employed 16S rRNA sequencing technology and liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based metabolic profiling combined with advanced differential and association analyses to investigate the correlation between the microbiome and the metabolome in male DSS rats. RESULTS A high-salt diet disrupted the balance of the intestinal flora and increased toxic metabolites (methyhistidines, creatinine, homocitrulline, and indoxyl sulfate), resulting in severe kidney damage. Canagliflozin contributed to reconstructing the intestinal flora of DSS rats by significantly increasing the abundance of Corynebacterium spp., Bifidobacterium spp., Facklamia spp., Lactobacillus spp., Ruminococcus spp., Blautia spp., Coprococcus spp., and Allobaculum spp. Moreover, the reconstruction of the intestinal microbiota led to significant changes in host amino acid metabolite concentrations. The concentration of uremic toxins, such as methyhistidines, creatinine, and homocitrulline, in the serum of rats was decreased by canagliflozin, which resulted in oxidative stress and renal injury alleviation. CONCLUSION Canagliflozin may change the production of metabolites and reduce the level of uremic toxins in the blood circulation by reconstructing the intestinal flora of DSS rats fed a high-salt diet, ultimately alleviating oxidative stress and renal injury.
Collapse
Affiliation(s)
- Lili He
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Qingjuan Zuo
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Sai Ma
- Department of Internal Medicine, Hebei General Hospital, Shijiazhuang, China
| | - Guorui Zhang
- Department of Cardiology, The Third Hospital of Shijiazhuang City Affiliated to Hebei Medical University, Shijiazhuang, China
| | - Zhongli Wang
- Department of Physical Examination Center, Hebei General Hospital, Shijiazhuang, China
| | - Tingting Zhang
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Jianlong Zhai
- Department of Cardiology, Hebei General Hospital, Shijiazhuang, China
| | - Yifang Guo
- Department of Geriatric Cardiology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
3
|
Abais-Battad JM, Dasinger JH, Lund H, Burns-Ray EC, Walton SD, Baldwin KE, Fehrenbach DJ, Cherian-Shaw M, Mattson DL. Sex-Dependency of T Cell-Induced Salt-Sensitive Hypertension and Kidney Damage. Hypertension 2024; 81:1511-1523. [PMID: 38757269 PMCID: PMC11168867 DOI: 10.1161/hypertensionaha.123.22608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND It is established that the immune system, namely T cells, plays a role in the development of hypertension and renal damage in male Dahl salt-sensitive (SS) rats, but far less is known about this relationship in females. Rats with genetically deleted T cells via CD247 gene mutation on the Dahl SS background (SSCD247-/-) were utilized to interrogate the effect of sex and T cells on salt sensitivity. METHODS We assessed the hypertensive and kidney injury phenotypes in male versus female SS and SSCD247-/- rats challenged with 3 weeks of high salt (4.0% NaCl). Differences in T cell activation genes were examined in renal T cells from male and female SS rats, and a sex-specific adoptive transfer was performed by injecting male or female splenocytes into either male or female SSCD247-/- recipients to determine the potential contribution of T cell sex. RESULTS The lack of functional T cells in SSCD247-/- rats significantly reduced salt-induced hypertension and proteinuria in both sexes, although SSCD247-/- females exhibited greater protection from kidney damage. Adoptive transfer of either Dahl SS male or female splenocytes into SSCD247-/- male recipients exacerbated hypertension and proteinuria compared with controls, while in SSCD247-/- female recipients, exacerbation of disease occurred only upon transfer of male, but not female, SS splenocytes. CONCLUSIONS The absence of T cells in the SSCD247-/- normalized sex differences in blood pressure, though sex differences in renal damage persisted. Splenocyte transfer experiments demonstrated that salt sensitivity is amplified if the sex of the T cell or the recipient is male.
Collapse
Affiliation(s)
| | - John Henry Dasinger
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | - Hayley Lund
- Department of Medicine, Medical College of Wisconsin, Milwaukee WI
| | - Emily C. Burns-Ray
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | - Samuel D. Walton
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | - Kaitlyn E. Baldwin
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | | | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| | - David L. Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta GA
| |
Collapse
|
4
|
Walton SD, Dasinger JH, Burns EC, Cherian-Shaw M, Abais-Battad JM, Mattson DL. Functional NADPH oxidase 2 in T cells amplifies salt-sensitive hypertension and associated renal damage. Am J Physiol Renal Physiol 2023; 325:F214-F223. [PMID: 37318993 PMCID: PMC10396224 DOI: 10.1152/ajprenal.00014.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023] Open
Abstract
Infiltrating T cells in the kidney amplify salt-sensitive (SS) hypertension and renal damage, but the mechanisms are not known. Genetic deletion of T cells (SSCD247-/-) or of the p67phox subunit of NADPH oxidase 2 (NOX2; SSp67phox-/-) attenuates SS hypertension in the Dahl SS rat. We hypothesized that reactive oxygen species produced by NOX2 in T cells drive the SS phenotype and renal damage. T cells were reconstituted by adoptively transferring splenocytes (∼10 million) from the Dahl SS (SS→CD247) rat, the SSp67phox-/- rat (p67phox→CD247), or only PBS (PBS→CD247) into the SSCD247-/- rat on postnatal day 5. Animals were instrumented with radiotelemeters and studied at 8 wk of age. There were no detectable differences in mean arterial pressure (MAP) or albuminuria between groups when rats were maintained on a low-salt (0.4% NaCl) diet. After 21 days of high-salt diet (4.0% NaCl), MAP and albuminuria were significantly greater in SS→CD247 rats compared with p67phox→CD247 and PBS→CD247 rats. Interestingly, there was no difference between p67phox→CD247 and PBS→CD247 rats in albuminuria or MAP after 21 days. The lack of CD3+ cells in PBS→CD247 rats and the presence of CD3+ cells in rats that received the T cell transfer demonstrated the effectiveness of the adoptive transfer. No differences in the number of CD3+, CD4+, or CD8+ cells were observed in the kidneys of SS→CD247 and p67phox→CD247 rats. These results indicate that reactive oxygen species produced by NOX2 in T cells participates in the amplification of SS hypertension and renal damage.NEW & NOTEWORTHY Our current work used the adoptive transfer of T cells that lack functional NADPH oxidase 2 into a genetically T cell-deficient Dahl salt-sensitive (SS) rat model. The results demonstrated that reactive oxygen species produced by NADPH oxidase 2 in T cells participate in the amplification of SS hypertension and associated renal damage and identifies a potential mechanism that exacerbates the salt-sensitive phenotype.
Collapse
Affiliation(s)
- Samuel D Walton
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - John Henry Dasinger
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Emily C Burns
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
5
|
McNally RJ, Morselli F, Farukh B, Chowienczyk PJ, Faconti L. A pilot study to evaluate the erythrocyte glycocalyx sensitivity to sodium as a marker for cellular salt sensitivity in hypertension. J Hum Hypertens 2023; 37:286-291. [PMID: 35414109 PMCID: PMC10063440 DOI: 10.1038/s41371-022-00683-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 03/02/2022] [Accepted: 03/25/2022] [Indexed: 11/09/2022]
Abstract
Supressed plasma renin in patients with primary hypertension is thought to be an indirect marker of sodium-induced volume expansion which is associated with more severe hypertension and hypertension-mediated organ damage. A novel test for erythrocyte glycocalyx sensitivity to sodium (eGCSS) has been proposed as a direct measure of sodium-induced damage on erythrocyte surfaces and a marker of sensitivity of the endothelium to salt in humans. Here we explore if eGCSS relates to plasma renin and other clinical and biochemical characteristics in a cohort of patients with primary hypertension. Hypertensive subjects (n = 85, 54% male) were characterised by blood biochemistry (including plasma renin/aldosterone), urine analysis for albumin-creatinine ratio (ACR), 24-h urine sodium/potassium excretion. eGCSS was measured using a commercially available kit. Correlations between eGCSS and clinical and biochemical characteristics were explored using Spearman's correlation coefficient and characteristics compared across tertiles of eGCSS. eGCSS was inversely correlated with renin (p < 0.05), with renin 17.72 ± 18 µU/l in the highest tertile of eGCSS compared to 84.27 ± 146.5 µU/l in the lowest (p = 0.012). eGCSS was positively correlated with ACR (p < 0.01), with ACR 7.37 ± 15.29 vs. 1.25 ± 1.52 g/mol for the highest vs. lowest tertiles of eGCSS (p < 0.05). eGCSS was not correlated with other clinical characteristics or biochemical measures. These results suggests that sodium retention in hypertension characterised by a low-renin state is associated with cell membrane damage reflected by eGCSS. This may contribute to the hypertension-mediated organ damage and the excess mortality associated with sodium overload and "salt sensitivity".
Collapse
Affiliation(s)
- Ryan J McNally
- King's College London, Department of Clinical Pharmacology, British Heart Foundation Centre, London, UK
| | - Franca Morselli
- King's College London, Department of Clinical Pharmacology, British Heart Foundation Centre, London, UK
| | - Bushra Farukh
- King's College London, Department of Clinical Pharmacology, British Heart Foundation Centre, London, UK
| | - Phil J Chowienczyk
- King's College London, Department of Clinical Pharmacology, British Heart Foundation Centre, London, UK
| | - Luca Faconti
- King's College London, Department of Clinical Pharmacology, British Heart Foundation Centre, London, UK.
| |
Collapse
|
6
|
Chivers JM, Whiles SA, Miles CB, Biederman BE, Ellison MF, Lovingood CW, Wright MH, Hoover DB, Raafey MA, Youngberg GA, Venkatachalam MA, Zheleznova NN, Yang C, Liu P, Kriegel AJ, Cowley AW, O'Connor PM, Picken MM, Polichnowski AJ. Brown-Norway chromosome 1 mitigates the upregulation of proinflammatory pathways in mTAL cells and subsequent age-related CKD in Dahl SS/JrHsdMcwi rats. Am J Physiol Renal Physiol 2023; 324:F193-F210. [PMID: 36475869 PMCID: PMC9886360 DOI: 10.1152/ajprenal.00145.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) has a strong genetic component; however, the underlying pathways are not well understood. Dahl salt-sensitive (SS)/Jr rats spontaneously develop CKD with age and are used to investigate the genetic determinants of CKD. However, there are currently several genetically diverse Dahl SS rats maintained at various institutions and the extent to which some exhibit age-related CKD is unclear. We assessed glomerulosclerosis (GS) and tubulointerstitial fibrosis (TIF) in 3- and 6-mo-old male and female SS/JrHsdMcwi, BN/NHsd/Mcwi [Brown-Norway (BN)], and consomic SS-Chr 1BN/Mcwi (SS.BN1) rats, in which chromosome 1 from the BN rat was introgressed into the genome of the SS/JrHsdMcwi rat. Rats were fed a 0.4% NaCl diet. GS (31 ± 3% vs. 7 ± 1%) and TIF (2.3 ± 0.2 vs. 0.5 ± 0.1) were significantly greater in 6-mo-old compared with 3-mo-old SS/JrHsdMcwi rats, and CKD was exacerbated in males. GS was minimal in 6- and 3-mo-old BN (3.9 ± 0.6% vs. 1.2 ± 0.4%) and SS.BN1 (2.4 ± 0.5% vs. 1.0 ± 0.3%) rats, and neither exhibited TIF. In SS/JrHsdMcwi and SS.BN1 rats, mean arterial blood pressure was significantly greater in 6-mo-old compared with 3-mo-old SS/JrHsdMcwi (162 ± 4 vs. 131 ± 2 mmHg) but not SS.BN1 (115 ± 2 vs. 116 ± 1 mmHg) rats. In 6-mo-old SS/JrHsdMcwi rats, blood pressure was significantly greater in females. RNA-sequencing analysis revealed that inflammatory pathways were upregulated in isolated medullary thick ascending tubules in 7-wk-old SS/JrHsdMcwi rats, before the development of tubule pathology, compared with SS.BN1 rats. In summary, SS/JrHsdMcwi rats exhibit robust age-related progression of medullary thick ascending limb abnormalities, CKD, and hypertension, and gene(s) on chromosome 1 have a major pathogenic role in such changes.NEW & NOTEWORTHY This study shows that the robust age-related progression of kidney disease in Dahl SS/JrHsdMcw rats maintained on a normal-salt diet is abolished in consomic SS.BN1 rats. Evidence that medullary thick ascending limb segments of SS/JrHsdMcw rats are structurally abnormal and enriched in proinflammatory pathways before the development of protein casts provides new insights into the pathogenesis of kidney disease in this model.
Collapse
Affiliation(s)
- Jacqueline M Chivers
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Shannon A Whiles
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Conor B Miles
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Brianna E Biederman
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Megan F Ellison
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Connor W Lovingood
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Marie H Wright
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
| | - Donald B Hoover
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| | - Muhammad A Raafey
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | - George A Youngberg
- Department of Pathology, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| | | | | | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Pengyuan Liu
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Paul M O'Connor
- Department of Physiology, Augusta University, Augusta, Georgia
| | - Maria M Picken
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Aaron J Polichnowski
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee
- Center of Excellence in Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|
7
|
Chu PL, Gigliotti JC, Cechova S, Bodonyi-Kovacs G, Wang YT, Chen L, Wassertheil-Smoller S, Cai J, Isakson BE, Franceschini N, Le TH. Collectrin ( Tmem27) deficiency in proximal tubules causes hypertension in mice and a TMEM27 variant associates with blood pressure in males in a Latino cohort. Am J Physiol Renal Physiol 2023; 324:F30-F42. [PMID: 36264884 PMCID: PMC9762972 DOI: 10.1152/ajprenal.00176.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/07/2022] [Accepted: 09/23/2022] [Indexed: 02/04/2023] Open
Abstract
Collectrin (Tmem27), an angiotensin-converting enzyme 2 homologue, is a chaperone of amino acid transporters in the kidney and endothelium. Global collectrin knockout (KO) mice have hypertension, endothelial dysfunction, exaggerated salt sensitivity, and diminished renal blood flow. This phenotype is associated with altered nitric oxide and superoxide balance and increased proximal tubule (PT) Na+/H+ exchanger isoform 3 (NHE3) expression. Collectrin is located on the X chromosome where genome-wide association population studies have largely been excluded. In the present study, we generated PT-specific collectrin KO (PT KO) mice to determine the precise contribution of PT collectrin in blood pressure homeostasis. We also examined the association of human TMEM27 single-nucleotide polymorphisms with blood pressure traits in 11,926 Hispanic Community Health Study/Study of Latinos (HCHS/SOL) Hispanic/Latino participants. PT KO mice exhibited hypertension, and this was associated with increased baseline NHE3 expression and diminished lithium excretion. However, PT KO mice did not display exaggerated salt sensitivity or a reduction in renal blood flow compared with control mice. Furthermore, PT KO mice exhibited enhanced endothelium-mediated dilation, suggesting a compensatory response to systemic hypertension induced by deficiency of collectrin in the PT. In HCHS/SOL participants, we observed sex-specific single-nucleotide polymorphism associations with diastolic blood pressure. In conclusion, loss of collectrin in the PT is sufficient to induce hypertension, at least in part, through activation of NHE3. Importantly, our model supports the notion that altered renal blood flow may be a determining factor for salt sensitivity. Further studies are needed to investigate the role of the TMEM27 locus on blood pressure and salt sensitivity in humans.NEW & NOTEWORTHY The findings of our study are significant in several ways: 1) loss of an amino acid chaperone in the proximal tubule is sufficient to cause hypertension, 2) the results in global and proximal tubule-specific collectrin knockout mice support the notion that vascular dysfunction is required for salt sensitivity or that impaired renal tubule function causes hypertension but is not sufficient to cause salt sensitivity, and 3) our study is the first to implicate a role of collectrin in human hypertension.
Collapse
Affiliation(s)
- Pei-Lun Chu
- Division of Nephrology, Fu Jen Catholic University Hospital, and School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Joseph C Gigliotti
- Department of Integrated Physiology and Pharmacology, Liberty University College of Osteopathic Medicine, Lynchburg, Virginia
| | - Sylvia Cechova
- Division of Nephrology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Gabor Bodonyi-Kovacs
- Division of Nephrology, Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Yves T Wang
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center Rochester, Rochester, New York
| | - Luojing Chen
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center Rochester, Rochester, New York
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center and Department of Molecular Physiology and Biophysics, University of Virginia Health System, Charlottesville, Virginia
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center Rochester, Rochester, New York
| |
Collapse
|
8
|
Maaliki D, Itani MM, Itani HA. Pathophysiology and genetics of salt-sensitive hypertension. Front Physiol 2022; 13:1001434. [PMID: 36176775 PMCID: PMC9513236 DOI: 10.3389/fphys.2022.1001434] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Most hypertensive cases are primary and heavily associated with modifiable risk factors like salt intake. Evidence suggests that even small reductions in salt consumption reduce blood pressure in all age groups. In that regard, the ACC/AHA described a distinct set of individuals who exhibit salt-sensitivity, regardless of their hypertensive status. Data has shown that salt-sensitivity is an independent risk factor for cardiovascular events and mortality. However, despite extensive research, the pathogenesis of salt-sensitive hypertension is still unclear and tremendously challenged by its multifactorial etiology, complicated genetic influences, and the unavailability of a diagnostic tool. So far, the important roles of the renin-angiotensin-aldosterone system, sympathetic nervous system, and immune system in the pathogenesis of salt-sensitive hypertension have been studied. In the first part of this review, we focus on how the systems mentioned above are aberrantly regulated in salt-sensitive hypertension. We follow this with an emphasis on genetic variants in those systems that are associated with and/or increase predisposition to salt-sensitivity in humans.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Maha M. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hana A. Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
9
|
Inverse salt sensitivity: an independent risk factor for cardiovascular damage in essential hypertension. J Hypertens 2022; 40:1504-1512. [PMID: 35881450 DOI: 10.1097/hjh.0000000000003174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Salt sensitivity is a powerful risk factor for cardiovascular (CV) disease and mortality in both normotensive and hypertensive patients. We investigated the predictive value of the salt sensitivity phenotype in the development of CV events and hypertensive target organ damage (TOD) among essential hypertensive patients. METHODS Eight hundred forty-four naive hypertensive patients were recruited and underwent an acute saline test during which blood pressure (BP) displayed either no substantial variation (salt-resistant, SR individuals), an increase (salt-sensitive, SS), or a paradoxical decrease (inverse salt-sensitive, ISS). Sixty-one patients with the longest monitored follow-up (median 16 years) for blood pressure and organ damage were selected for the present study. A clinical score for TOD development based on the severity and the age of onset was set up by considering hypertensive heart disease, cerebrovascular damage, microalbuminuria, and vascular events. RESULTS CV events were significantly higher among SS and ISS than in SR patients. The relative risk of developing CV events was 12.67 times higher in SS than SR and 5.94 times higher in ISS than SR patients. The development of moderate to severe TOD was 10-fold higher in SS and over 15-fold higher in ISS than in SR patients. Among the three phenotypes, changes in plasma endogenous ouabain were linked with the blood pressure effects of saline. CONCLUSIONS Salt sensitivity and inverse salt sensitivity appear to be equivalent risk factors for CV events. The response to an acute saline test is predictive of CV damage for newly identified ISS individuals.
Collapse
|
10
|
Salt-Sensitive Ileal Microbiota Plays a Role in Atrial Natriuretic Peptide Deficiency-Induced Cardiac Injury. Nutrients 2022; 14:nu14153129. [PMID: 35956306 PMCID: PMC9370783 DOI: 10.3390/nu14153129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Atrial natriuretic peptide (ANP) activity deficiency contributes to salt-sensitive hypertension in humans and mice. However, the role of ileal microbiota in salt sensitivity in ANP deficiency-related cardiac injury has not been investigated yet. This study used ANP−/− mice to analyze the role of the salt-sensitive ileal microbiome on cardiac injury. ANP−/− mice showed an increase in blood pressure (BP), the heart weight/body weight (HW/BW) ratio, and cardiac hypertrophy compared with wild-type (WT) mice. ANP deficiency did not impact the histological structure but reduced occludin expression in the ileum. Antibiotics significantly relieved BP and cardiac hypertrophy in ANP−/− mice. A high-salt diet (HSD) increased BP, the HW/BW ratio, and cardiac hypertrophy/fibrosis in WT and ANP−/− mice, and an HSD treatment in ANP−/− mice exacerbated these cardiac parameters. The HSD markedly decreased muscularis layer thickening, villus length, and numbers of Paneth and goblet cells in the ileum of WT and ANP−/− mice. Furthermore, the HSD increased the level of TLR4 and IL-1β in ANP−/− mice ileum compared with WT mice. Antibiotics reduced the HW/BW ratio, cardiac hypertrophy/fibrosis, and the level of TLR4 and IL-1β in the ileum, and rescued the muscularis layer thickening, villus length, and numbers of Paneth and goblet cells in the ileum of HSD-ANP−/− mice. Importantly, ANP deficiency induced the colonization of Burkholderiales bacterium YL45, Lactobacillus johnsonii, and Lactobacillus reuteri in the ileum on the NSD diet, which was only observed in HSD-induced WT mice but not in WT mice on the NSD. Besides, the HSD significantly enhanced the sum of the percentage of the colonization of Burkholderiales bacterium YL45, Lactobacillus johnsonii, and Lactobacillus reuteri in the ileum of ANP−/− mice. Ileal microbiota transfer (IMT) from ANP−/− mice to healthy C57BL/6J mice drove Lactobacillus johnsonii and Lactobacillus reuteri colonization in the ileum, which manifested an increase in BP, the HW/BW ratio, cardiac hypertrophy, and ileal pathology compared with IMT from WT mice. The HSD in C57BL/6J mice with IMT from ANP−/− mice drove the colonization of Burkholderiales bacterium YL45, Lactobacillus johnsonii, and Lactobacillus reuteri in the ileum and further exacerbated the cardiac and ileal pathology. Our results suggest that salt-sensitive ileal microbiota is probably related to ANP deficiency-induced cardiac injury.
Collapse
|
11
|
Kurtz T, Pravenec M, DiCarlo S. Mechanism-based strategies to prevent salt sensitivity and salt-induced hypertension. Clin Sci (Lond) 2022; 136:599-620. [PMID: 35452099 PMCID: PMC9069470 DOI: 10.1042/cs20210566] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/15/2022]
Abstract
High-salt diets are a major cause of hypertension and cardiovascular (CV) disease. Many governments are interested in using food salt reduction programs to reduce the risk for salt-induced increases in blood pressure and CV events. It is assumed that reducing the salt concentration of processed foods will substantially reduce mean salt intake in the general population. However, contrary to expectations, reducing the sodium density of nearly all foods consumed in England by 21% had little or no effect on salt intake in the general population. This may be due to the fact that in England, as in other countries including the U.S.A., mean salt intake is already close to the lower normal physiologic limit for mean salt intake of free-living populations. Thus, mechanism-based strategies for preventing salt-induced increases in blood pressure that do not solely depend on reducing salt intake merit attention. It is now recognized that the initiation of salt-induced increases in blood pressure often involves a combination of normal increases in sodium balance, blood volume and cardiac output together with abnormal vascular resistance responses to increased salt intake. Therefore, preventing either the normal increases in sodium balance and cardiac output, or the abnormal vascular resistance responses to salt, can prevent salt-induced increases in blood pressure. Suboptimal nutrient intake is a common cause of the hemodynamic disturbances mediating salt-induced hypertension. Accordingly, efforts to identify and correct the nutrient deficiencies that promote salt sensitivity hold promise for decreasing population risk of salt-induced hypertension without requiring reductions in salt intake.
Collapse
Affiliation(s)
- Theodore W. Kurtz
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94017-0134, U.S.A
| | - Michal Pravenec
- Institute of Physiology, Czech Academy of Sciences, Prague 14220, Czech Republic
| | - Stephen E. DiCarlo
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, U.S.A
| |
Collapse
|
12
|
Damianaki A, Polychronopoulou E, Wuerzner G, Burnier M. New Aspects in the Management of Hypertension in Patients with Chronic Kidney Disease not on Renal Replacement Therapy. High Blood Press Cardiovasc Prev 2021; 29:125-135. [PMID: 34910287 PMCID: PMC8942929 DOI: 10.1007/s40292-021-00495-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/30/2021] [Indexed: 11/07/2022] Open
Abstract
With chronic kidney disease (CKD) being a global arising health problem, strategies for delaying kidney disease progression and reducing the high cardiovascular risk inherent to CKD, are the main objectives of the actual management of patients with kidney diseases. In these patients, the control of arterial hypertension is essential, as high blood pressure (BP) is a strong determinant of worst cardiovascular and renal outcomes. Achieving target blood pressures recommended by international guidelines is mandatory and often demands a multiple levels management, including several pharmacological and lifestyle measures. Even in the presence of adequate BP control, the residual cardiovascular risk remains high. In this respect, the recent demonstration that novel agents such as sodium glucose transporter 2 (SGLT2) inhibitors or the new non-steroidal mineralocorticoid antagonist finerenone can retard the progression of kidney diseases and reduce cardiovascular mortality on top of standard of care treatment with renin-angiotensin system inhibitors represent enormous progresses. These studies also demonstrate that cardiovascular and renal protection can be obtained beyond blood pressure control. Other promising novelties are still to come such as renal denervation and endothelin receptor antagonists in the setting of diabetic and non-diabetic kidney diseases. In the present review, we shall discuss the classic and the new aspects for the management of hypertension in CKD, integrating the new data from recent clinical studies.
Collapse
Affiliation(s)
- Aikaterini Damianaki
- Service of Nephrology and Hypertension, University Hospital, Rue du Bugnon 17, 1011, Lausanne, Switzerland
| | - Erietta Polychronopoulou
- Service of Nephrology and Hypertension, University Hospital, Rue du Bugnon 17, 1011, Lausanne, Switzerland
| | - Gregoire Wuerzner
- Service of Nephrology and Hypertension, University Hospital, Rue du Bugnon 17, 1011, Lausanne, Switzerland.,Hypertension Research Foundation, Saint-Légier, Switzerland
| | - Michel Burnier
- Service of Nephrology and Hypertension, University Hospital, Rue du Bugnon 17, 1011, Lausanne, Switzerland. .,Hypertension Research Foundation, Saint-Légier, Switzerland.
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW In this article, we summarize the current literature supporting metabolic and redox signaling pathways as important mechanisms underlying T cell activation in the context of hypertension. RECENT FINDINGS T cell immunometabolism undergoes dramatic remodeling in order to meet the demands of T cell activation, differentiation, and proliferation. Recent evidence demonstrates that the T cell oxidation-reduction (redox) system also undergoes significant changes upon activation, which can itself modulate metabolic processes and T cell function. Dysregulation of these signaling pathways can lead to aberrant T cell activation and inappropriate ROS production, both of which are linked to pathological conditions like hypertension. While the contribution of T cells to the progression of hypertension has been thoroughly investigated, how T cell metabolism and redox signaling changes, both separately and together, is an area of study that remains largely untouched. This review presents evidence from our own laboratory as well as others to highlight the importance of these two mechanisms in the study of hypertension.
Collapse
|
14
|
Ma Z, Hummel SL, Sun N, Chen Y. From salt to hypertension, what is missed? J Clin Hypertens (Greenwich) 2021; 23:2033-2041. [PMID: 34846798 PMCID: PMC8696232 DOI: 10.1111/jch.14402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/20/2021] [Accepted: 11/03/2021] [Indexed: 01/18/2023]
Abstract
Excess salt intake is viewed as a major contributor to hypertension and cardiovascular disease, and dietary salt restriction is broadly recommended by public health guidelines. However, individuals can have widely varying physiological responses to salt intake, and a tailored approach to evaluation and intervention may be needed. The traditional sodium related concepts are challenging to assess clinically for two reasons: (1) spot and 24-hour urine sodium are frequently used to evaluate salt intake, but are more suitable for population study, and (2) some adverse effects of salt may be blood pressure-independent. In recent years, previously unknown mechanisms of sodium absorption and storage have been discovered. This review will outline the limitations of current methods to assess sodium balance and discuss new potential evaluation methods and treatment targets.
Collapse
Affiliation(s)
- Zhiyi Ma
- Cardiology DepartmentBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
| | - Scott L. Hummel
- Ann Arbor Veterans Affairs Health SystemUniversity of Michigan Frankel Cardiovascular CenterAnn ArborMichiganUSA
| | - Ningling Sun
- Cardiology DepartmentHeart CenterPeking University People's HospitalBeijingChina
| | - Yuanyuan Chen
- Cardiology DepartmentHeart CenterPeking University People's HospitalBeijingChina
| |
Collapse
|
15
|
Abstract
High salt intake is associated with hypertension, which is a leading modifiable risk factor for cardiovascular disease (CVD) and chronic kidney disease (CKD). International Guidelines recommend a large reduction in the consumption of sodium to reduce blood pressure, organ damage, and mortality. In its early stages, the symptoms of CKD are generally not apparent. CKD proceeds in a "silent" manner, necessitating the need for urinary biomarkers to detect kidney damage at an early stage. Since traditional renal biomarkers, such as serum creatinine, are not sufficiently sensitive, difficulties are associated with detecting kidney damage induced by a high salt intake, particularly in normotensive individuals. Several new biomarkers for renal tubular damage, such as neutrophil gelatinase-associated lipocalin (NGAL), kidney injury molecule-1 (KIM-1), vanin-1, liver-type fatty acid-binding protein (L-FABP), and monocyte chemotactic protein-1 (MCP-1), have recently been identified. However, few studies have investigated early biomarkers for CKD progression associated with a high salt diet. This chapter provides insights into novel biomarkers for CKD in normo- and hypertensive individuals with a high salt intake. Recent studies using spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto rats (WKY) fed a high salt diet identified urinary vanin-1 and NGAL as early biomarkers for renal tubular damage in SHR and WKY, whereas urinary KIM-1 was a useful biomarker for salt-induced renal injury in SHR only. Clinical studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Keiko Hosohata
- Education and Research Center for Clinical Pharmacy, Osaka University of Pharmaceutical Sciences, Takatsuki, Osaka, Japan.
| |
Collapse
|
16
|
Tagawa K, Tsuru Y, Yokoi K, Aonuma T, Hashimoto J. Albuminuria Intensifies the Link Between Urinary Sodium Excretion and Central Pulse Pressure in the General Population: The Wakuya Study. Am J Hypertens 2021; 34:851-857. [PMID: 33893813 PMCID: PMC8385571 DOI: 10.1093/ajh/hpab057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/23/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Central pulse pressure (cPP) is responsible for the hemodynamics of vital organs, and monitoring this parameter is important for cardiovascular disease (CVD) prevention. Excess sodium intake and (micro)albuminuria (a manifestation of renal microvascular damage) are known to be strong predictors of CVD. We sought to investigate the cross-sectional relationships among dietary sodium intake, albuminuria, and cPP in a general population cohort. METHODS The subjects were 933 apparently healthy adults (mean age, 56 ± 10 years). Radial pressure waveforms were recorded with applanation tonometry to estimate mean arterial pressure (MAP), cPP, forward and backward pressure amplitudes, and augmentation index. The urinary sodium/creatinine and albumin/creatinine ratios were measured in spot urine samples. RESULTS Both the urinary sodium/creatinine and albumin/creatinine ratios were positively correlated with cPP, even after adjusting for MAP (P < 0.001). Moreover, both ratios had a synergistic influence on increasing the cPP independent of age, sex, estimated glomerular filtration rate, hyperlipidemia, and diabetes (interaction P = 0.04). A similar synergistic influence was found on the forward pressure amplitude, but not on the backward pressure amplitude or augmentation index. The overall results were not altered when the urinary albumin/creatinine ratio was replaced with the existence of chronic kidney disease (CKD). CONCLUSIONS (Micro)albuminuria strengthens the positive association between urinary sodium excretion and cPP and systolic forward pressure. Excess sodium intake may magnify the cardiovascular risk by widening the aortic pulsatile pressure, particularly in the presence of concomitant CKD.
Collapse
Affiliation(s)
- Kaname Tagawa
- Miyagi University of Education Medical Center, Sendai, Japan
| | - Yusuke Tsuru
- Wakuya National Health Insurance Hospital, Miyagi, Japan
| | - Katsumi Yokoi
- Wakuya National Health Insurance Hospital, Miyagi, Japan
| | | | | |
Collapse
|
17
|
Abais-Battad JM, Saravia FL, Lund H, Dasinger JH, Fehrenbach DJ, Alsheikh AJ, Zemaj J, Kirby JR, Mattson DL. Dietary influences on the Dahl SS rat gut microbiota and its effects on salt-sensitive hypertension and renal damage. Acta Physiol (Oxf) 2021; 232:e13662. [PMID: 33866692 PMCID: PMC9835005 DOI: 10.1111/apha.13662] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/07/2021] [Accepted: 04/11/2021] [Indexed: 01/14/2023]
Abstract
AIM Our previous studies have demonstrated the importance of dietary factors in the determination of hypertension in Dahl salt-sensitive (SS) rats. Since the gut microbiota has been implicated in chronic diseases like hypertension, we hypothesized that dietary alterations shift the microbiota to mediate the development of salt-sensitive hypertension and renal disease. METHODS This study utilized SS rats from the Medical College of Wisconsin (SS/MCW) maintained on a purified, casein-based diet (0.4% NaCl AIN-76A, Dyets) and from Charles River Laboratories (SS/CRL) fed a whole grain diet (0.75% NaCl 5L79, LabDiet). Faecal 16S rDNA sequencing was used to phenotype the gut microbiota. Directly examining the contribution of the gut microbiota, SS/CRL rats were administered faecal microbiota transfer (FMT) experiments with either SS/MCW stool or vehicle (Vehl) in conjunction with the HS AIN-76A diet. RESULTS SS/MCW rats exhibit renal damage and inflammation when fed high salt (HS, 4.0% NaCl AIN-76A), which is significantly attenuated in SS/CRL. Gut microbiota phenotyping revealed distinct profiles that correlate with disease severity. SS/MCW FMT worsened the SS/CRL response to HS, evidenced by increased albuminuria (67.4 ± 6.9 vs 113.7 ± 25.0 mg/day, Vehl vs FMT, P = .007), systolic arterial pressure (158.6 ± 5.8 vs 177.8 ± 8.9 mmHg, Vehl vs FMT, P = .09) and renal T-cell infiltration (1.9-fold). Amplicon sequence variant (ASV)-based analysis of faecal 16S rDNA sequencing data revealed taxa that significantly shifted with FMT: Erysipelotrichaceae_2, Parabacteroides gordonii, Streptococcus alactolyticus, Bacteroidales_1, Desulfovibrionaceae_2, Ruminococcus albus. CONCLUSIONS These data demonstrate that dietary modulation of the gut microbiota directly contributes to the development of Dahl SS hypertension and renal injury.
Collapse
Affiliation(s)
- Justine M. Abais-Battad
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Fatima L. Saravia
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hayley Lund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John Henry Dasinger
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Daniel J. Fehrenbach
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ammar J. Alsheikh
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeylan Zemaj
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - John R. Kirby
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David L. Mattson
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
18
|
Trujillo H, Caravaca-Fontán F, Caro J, Morales E, Praga M. The Forgotten Antiproteinuric Properties of Diuretics. Am J Nephrol 2021; 52:435-449. [PMID: 34233330 DOI: 10.1159/000517020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Although diuretics are one of the most widely used drugs by nephrologists, their antiproteinuric properties are not generally taken into consideration. SUMMARY Thiazide diuretics have been shown to reduce proteinuria by >35% in several prospective controlled studies, and these values are markedly increased when combined with a low-salt diet. Thiazide-like diuretics (indapamide and chlorthalidone) have shown similar effectiveness. The antiproteinuric effect of mineralocorticoid receptor antagonists (spironolactone, eplerenone, and finerenone) has been clearly established through prospective and controlled studies, and treatment with finerenone reduces the risk of chronic kidney disease progression in type-2 diabetic patients. The efficacy of other diuretics such as amiloride, triamterene, acetazolamide, or loop diuretics has been less explored, but different investigations suggest that they might share the same antiproteinuric properties of other diuretics that should be evaluated through controlled studies. Although the inclusion of sodium-glucose cotransporter-2 inhibitors (SGLT2i) among diuretics is a controversial issue, their renoprotective and cardioprotective properties, confirmed in various landmark trials, constitute a true revolution in the treatment of patients with kidney disease. Recent subanalyses of these trials have shown that the early antiproteinuric effect induced by SGLT2i predicts long-term preservation of kidney function. Key Message: Whether the early reduction in proteinuria induced by diuretics other than finerenone and SGLT2i, as summarized in this review, also translates into long-term renoprotection requires further prospective and observational studies. In any case, it is important for the clinician to be aware of the antiproteinuric properties of drugs so often used in daily clinical practice.
Collapse
Affiliation(s)
- Hernando Trujillo
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain,
| | | | - Jara Caro
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
| | - Enrique Morales
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Praga
- Department of Nephrology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Madrid, Spain
- Department of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
19
|
Burnier M. Sodium intake and progression of chronic kidney disease-has the time finally come to do the impossible: a prospective randomized controlled trial? Nephrol Dial Transplant 2021; 36:381-384. [PMID: 32980869 DOI: 10.1093/ndt/gfaa120] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Indexed: 12/13/2022] Open
Affiliation(s)
- Michel Burnier
- Service of Nephrology and Hypertension, University Hospital, Lausanne, Switzerland.,Hypertension Research Foundation, St-Légier, Switzerland
| |
Collapse
|
20
|
Mattson DL, Dasinger JH, Abais-Battad JM. Amplification of Salt-Sensitive Hypertension and Kidney Damage by Immune Mechanisms. Am J Hypertens 2021; 34:3-14. [PMID: 32725162 PMCID: PMC7891248 DOI: 10.1093/ajh/hpaa124] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/27/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
Humans with salt-sensitive (SS) hypertension demonstrate increased morbidity, increased mortality, and renal end-organ damage when compared with normotensive subjects or those with salt-resistant hypertension. Increasing evidence indicates that immune mechanisms play an important role in the full development of SS hypertension and associated renal damage. Recent experimental advances and studies in animal models have permitted a greater understanding of the mechanisms of activation and action of immunity in this disease process. Evidence favors a role of both innate and adaptive immune mechanisms that are triggered by initial, immune-independent alterations in blood pressure, sympathetic activity, or tissue damage. Activation of immunity, which can be enhanced by a high-salt intake or by alterations in other components of the diet, leads to the release of cytokines, free radicals, or other factors that amplify renal damage and hypertension and mediate malignant disease.
Collapse
Affiliation(s)
- David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - John Henry Dasinger
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
21
|
Polina I, Spicer MJ, Domondon M, Schibalski RS, Sarsenova E, Sultanova RF, Ilatovskaya DV. Inhibition of neprilysin with sacubitril without RAS blockage aggravates renal disease in Dahl SS rats. Ren Fail 2021; 43:315-324. [PMID: 33541194 PMCID: PMC8901277 DOI: 10.1080/0886022x.2021.1879856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Salt-sensitive (SS) hypertension is accompanied with severe cardiorenal complications. In this condition, elevated blood pressure (BP) resulting from salt retention is associated with counterintuitively lower levels of atrial natriuretic peptide (ANP). In plasma, ANP is degraded by the neprilysin; therefore, pharmacological inhibition of this metalloprotease (i.e., with sacubitril) can be employed to increase ANP level. We have shown earlier that sacubitril in combination with valsartan (75 μg/day each) had beneficial effects on renal function in Dahl SS rats. The goal of this study was to evaluate the effects of a higher dose of sacubitril on renal damage in this model. To induce hypertension, male Dahl SS rats were fed a 4% NaCl diet (HS) for 21 days, and were administered sacubitril (125 μg/day) or vehicle via s.c. osmotic pumps. At the end of the HS challenge, both groups exhibited similar outcomes for GFR, heart weight, plasma electrolytes, BUN, and creatinine. Sacubitril exacerbated kidney hypertrophy, but did not affect levels of renal fibrosis. We also observed aggravated glomerular lesions and increased formation of protein casts in the sacubitril-treated animals compared to controls. Thus, in Dahl SS rats, administration of sacubitril without renin-angiotensin-system blockage had adverse effects on renal disease progression, particularly in regards to glomerular damage and protein cast formation. We can speculate that while ANP levels are increased because of neprilysin inhibition, there are off-target effects of sacubitril, which are detrimental to renal function in the SS hypertensive state.
Collapse
Affiliation(s)
- Iuliia Polina
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, USA
| | - Morgan J Spicer
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, USA
| | - Mark Domondon
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, USA
| | - Ryan S Schibalski
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, USA
| | - Elizaveta Sarsenova
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, USA.,Saint-Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | - Regina F Sultanova
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, USA.,Saint-Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | - Daria V Ilatovskaya
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
22
|
Andersen H, Hansen MH, Buhl KB, Stæhr M, Friis UG, Enggaard C, Supramaniyam S, Lund IK, Svenningsen P, Hansen PBL, Jensen BL. Plasminogen Deficiency and Amiloride Mitigate Angiotensin II-Induced Hypertension in Type 1 Diabetic Mice Suggesting Effects Through the Epithelial Sodium Channel. J Am Heart Assoc 2020; 9:e016387. [PMID: 33215566 PMCID: PMC7763785 DOI: 10.1161/jaha.120.016387] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Diabetic nephropathy is a common diabetes mellitus complication associated with hypertension, proteinuria, and excretion of urinary plasmin that activates the epithelial sodium channel, ENaC, in vitro. Here we hypothesized that the deletion of plasminogen and amiloride treatment protect against hypertension in diabetes mellitus. Methods and Results Male plasminogen knockout (plasminogen-deficient [Plg-/-]) and wild-type mice were rendered diabetic with streptozotocin. Arterial blood pressure was recorded continuously by indwelling catheters before and during 10 days of angiotensin II infusion (ANGII; 30-60 ng/kg per minute). The effect of amiloride infusion (2 mg/kg per day, 4 days) was tested in wild-type, diabetic ANGII-treated mice. Streptozotocin increased plasma and urine glucose concentrations and 24-hour urine albumin and plasminogen excretion. Diabetic Plg-/- mice displayed larger baseline albuminuria and absence of urine plasminogen. Baseline mean arterial blood pressure did not differ between groups. Although ANGII elevated blood pressure in wild-type, diabetic wild-type, and Plg-/- control mice, ANGII did not change blood pressure in diabetic Plg-/- mice. Compared with ANGII infusion alone, wild-type ANGII-infused diabetic mice showed blood pressure reduction upon amiloride treatment. There was no difference in plasma renin, ANGII, aldosterone, tissue prorenin receptor, renal inflammation, and fibrosis between groups. Urine from wild-type mice evoked larger amiloride-sensitive current than urine from Plg-/- mice with or without diabetes mellitus. Full-length γ-ENaC and α-ENaC subunit abundances were not changed in kidney homogenates, but the 70 kDa γ-ENaC cleavage product was increased in diabetic versus nondiabetic mice. Conclusions Plasmin promotes hypertension in diabetes mellitus with albuminuria likely through the epithelial sodium channel.
Collapse
Affiliation(s)
- Henrik Andersen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Maria Høj Hansen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Kristian B Buhl
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Mette Stæhr
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Ulla G Friis
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Camilla Enggaard
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Shanya Supramaniyam
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Ida K Lund
- The Finsen Laboratory Copenhagen University Hospital, and Biotech Research and Innovation Center (BRIC) University of Copenhagen Denmark
| | - Per Svenningsen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| | - Pernille B L Hansen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark.,Research and Early Development Cardiovascular, Renal and Metabolism BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense Denmark
| |
Collapse
|
23
|
Wang W, Chonchol M, Seals DR, Nowak KL. Dietary sodium restriction decreases urinary NGAL in older adults with moderately elevated systolic blood pressure free from chronic kidney disease. J Investig Med 2020; 68:1271-1275. [PMID: 32699180 PMCID: PMC7531580 DOI: 10.1136/jim-2020-001343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2020] [Indexed: 01/22/2023]
Abstract
Increased aortic stiffness may contribute to kidney damage by transferring excessive flow pulsatility to susceptible renal microvasculature, leading to constriction or vessel loss. We previously demonstrated that 5 weeks of dietary sodium restriction (DSR) reduces large-elastic artery stiffness as well as blood pressure in healthy middle-aged/older adults with moderately elevated systolic blood pressure (SBP) who are free from chronic kidney disease (CKD). We hypothesized that DSR in this cohort would also reduce urinary concentrations of renal tubular injury biomarkers, which predict incident CKD in the general population. We performed a post hoc analysis using stored 24 hours urine samples collected in 13 participants as part of a randomized, double-blind, crossover clinical trial of DSR (low sodium (LS) target: 50 mmol/day; normal sodium (NS) target: 150 mmol/day). Participants were 61±2 (mean±SEM) years (8 M/5 F) with a baseline blood pressure of 139±2/82±2 mm Hg and an estimated glomerular filtration rate of 79±3 mL/min/1.73 m2 Twenty-four hour urinary sodium excretion was reduced from 149±7 to 66±8 mmol/day during week 5. Despite having preserved kidney function, participants had a 31% reduction in urinary neutrophil gelatinase-associated lipocalin concentrations with just 5 weeks of DSR (LS: 2.8±0.6 vs NS: 4.2±0.8 ng/mL, p<0.05). Results were similar when normalized to urinary creatinine (urinary creatinine did not change between conditions). Concentrations of another kidney tubular injury biomarker, kidney injury molecule-1, were below the detectable limit in all but one sample. In conclusion, DSR reduces an established clinical biomarker of kidney tubular damage in adults with moderately elevated SBP who are free from prevalent kidney disease.
Collapse
Affiliation(s)
- Wei Wang
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, USA
| | - Kristen L Nowak
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
24
|
Naser AM, Rahman M, Unicomb L, Doza S, Selim S, Chaity M, Luby SP, Anand S, Staimez L, Clasen TF, Gujral UP, Gribble MO, Narayan KMV. Past Sodium Intake, Contemporary Sodium Intake, and Cardiometabolic Health in Southwest Coastal Bangladesh. J Am Heart Assoc 2020; 9:e014978. [PMID: 32875927 PMCID: PMC7727005 DOI: 10.1161/jaha.119.014978] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 07/30/2020] [Indexed: 02/07/2023]
Abstract
Background We compared the relationship of past and contemporary sodium (Na) intake with cardiometabolic biomarkers. Methods and Results A total of 1191 participants' data from a randomized controlled trial in coastal Bangladesh were analyzed. Participants provided 24-hour urine Na (24UNa) data for 5 monthly visits. Their fasting blood glucose, total cholesterol, triglycerides, high-density lipoprotein, blood pressure, and 24-hour urine protein were measured at the fifth visit. Participants' mean 24UNa over the first 4 visits was the past Na, and 24UNa of the fifth visit was the contemporary Na intake. We estimated the prevalence ratios of elevated cardiometabolic biomarkers and metabolic syndrome across 24UNa tertiles by multilevel logistic regression using participant-, household-, and community-level random intercepts. Models were adjusted for age, sex, body mass index, smoking, physical activity, alcohol consumption, sleep hours, religion, and household wealth. Compared with participants in tertile 1 of past urine Na, those in tertile 3 had 1.46 (95% CI, 1.08-1.99) times higher prevalence of prediabetes or diabetes mellitus, 5.49 (95% CI, 2.73-11.01) times higher prevalence of large waist circumference, and 1.60 (95% CI, 1.04-2.46) times higher prevalence of metabolic syndrome. Compared with participants in tertile 1 of contemporary urine Na, those in tertile 3 had 1.93 (95% CI, 1.24-3.00) times higher prevalence of prediabetes or diabetes mellitus, 3.14 (95% CI, 1.45-6.83) times higher prevalence of proteinuria, and 2.23 (95% CI, 1.34-3.71) times higher prevalence of large waist circumference. Conclusions Both past and contemporary Na intakes were associated with higher cardiometabolic disease risk.
Collapse
Affiliation(s)
- Abu Mohd Naser
- Emory Global Diabetes Research CenterHubert Department of Global HealthRollins School of Public HealthEmory UniversityAtlantaGA
| | - Mahbubur Rahman
- International Centre for Diarrhoeal Disease ResearchBangladesh (icddr,b)DhakaBangladesh
| | - Leanne Unicomb
- International Centre for Diarrhoeal Disease ResearchBangladesh (icddr,b)DhakaBangladesh
| | - Solaiman Doza
- International Centre for Diarrhoeal Disease ResearchBangladesh (icddr,b)DhakaBangladesh
| | - Shahjada Selim
- Department of EndocrinologyBangabandhu Sheikh Mujib Medical UniversityDhakaBangladesh
| | | | - Stephen P. Luby
- Division of Infectious Diseases and Geographic MedicineStanford UniversityStanfordCA
| | - Shuchi Anand
- Division of NephrologySchool of MedicineStanford UniversityStanfordCA
| | - Lisa Staimez
- Emory Global Diabetes Research CenterHubert Department of Global HealthRollins School of Public HealthEmory UniversityAtlantaGA
| | - Thomas F. Clasen
- Gangarosa Department of Environmental Health SciencesRollins School of Public HealthEmory UniversityAtlantaGA
| | - Unjali P. Gujral
- Emory Global Diabetes Research CenterHubert Department of Global HealthRollins School of Public HealthEmory UniversityAtlantaGA
| | - Matthew O. Gribble
- Gangarosa Department of Environmental Health SciencesRollins School of Public HealthEmory UniversityAtlantaGA
- Department of EpidemiologyRollins School of Public HealthEmory UniversityAtlantaGA
| | - K. M. Venkat Narayan
- Emory Global Diabetes Research CenterHubert Department of Global HealthRollins School of Public HealthEmory UniversityAtlantaGA
| |
Collapse
|
25
|
Mehrvar S, Foomani FH, Shimada S, Yang C, Zheleznova NN, Mostaghimi S, Cowley AW, Ranji M. The early effects of uninephrectomy on rat kidney metabolic state using optical imaging. JOURNAL OF BIOPHOTONICS 2020; 13:e202000089. [PMID: 32436651 DOI: 10.1002/jbio.202000089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 05/09/2023]
Abstract
Uninephrectomy (UNX) is known to result in structural and metabolic changes to the remaining kidney, although it is uncertain if this alters the mitochondrial redox state and how soon such changes may occur. A custom-designed fluorescence cryo-imaging technique was used to quantitatively assess the effect of UNX by measuring the levels of nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FAD) in the remaining kidney. Kidneys were snap-frozen 3 days following UNX, and the intrinsic fluorescence of NADH and FAD were optically acquired. The 3D images were created to characterize the NADH/FAD redox ratios (RR) of the right kidneys, which underwent UNX and the remaining kidneys 3 days following UNX. Both the NADPH-oxidases (Nox2 and Nox4) and the mitochondria are the main sources of reactive oxygen species (ROS) production in tubular epithelial cells. Responses to the UNX were obtained in kidneys of normal Sprague Dawley (SD) rats, Dahl salt-sensitive (SS) rats and SS rats in which NADPH-oxidase isoform 4 (Nox4) was knocked out (SSNox4-/- ). The results found that each of the strains exhibited similar increase in kidney weights averaging 17% after 3 days of UNX. SD and SSNox4-/- rats both exhibited global reductions of the RR (P < .05) with a similar tendency observed in SS rats (P < .08), indicating increased ROS production. The unexpected reduction of the RR in the remnant kidneys of SSNox4-/- rats indicates that mechanisms independent of H2 O2 produced from Nox4 may be responsible for this global increase of ROS. We propose that the reduced RR was largely a consequence of enhanced mitochondrial bioenergetics due to increased tubular workload of the remaining kidney. The data indicate that mitochondria become the dominant source of increased ROS following UNX and could represent an important hypertrophic signaling mechanism.
Collapse
Affiliation(s)
- Shima Mehrvar
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Farnaz H Foomani
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Satoshi Shimada
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | | | - Soudeh Mostaghimi
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin, USA
| | - Mahsa Ranji
- Biophotonics Lab, Department of Electrical Engineering, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| |
Collapse
|
26
|
Yoshimura R, Yamamoto R, Shinzawa M, Tomi R, Ozaki S, Fujii Y, Ito T, Tanabe K, Moriguchi Y, Isaka Y, Moriyama T. Frequency of alcohol drinking modifies the association between salt intake and albuminuria: a 1-year observational study. Hypertens Res 2020; 43:1249-1256. [PMID: 32651558 DOI: 10.1038/s41440-020-0503-4] [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: 03/21/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/16/2022]
Abstract
Albuminuria is an important risk factor for end-stage kidney disease and cardiovascular mortality. This 1-year observational study aimed to assess the effect modification of alcohol drinking on the association between salt intake and albuminuria. Overall, 448 employees at a pharmaceutical company in Japan who underwent annual health checkups in both 2017 and 2018 were evaluated. The main exposure of interest was drinking frequency at their first checkups categorized as rarely, occasionally, and daily. To assess the association between the changes in salt intake and albuminuria, the differences in salt intake estimated from single-spot urine specimens and the urinary albumin-to-creatinine ratio (UACR) between 2017 and 2018 were calculated for each subject. A multivariable-adjusted linear regression model showed a significant association between ∆salt intake and ∆Log UACR (per 1 g/day of ∆salt intake, adjusted ß 0.16 [95% confidence interval 0.14, 0.19]) and an effect modification between drinking frequency and ∆salt intake (P for interaction = 0.088). The association between ∆salt intake and ∆Log UACR was enhanced by drinking frequency in a dose-dependent manner (per 1 g/day of ∆salt intake, adjusted ß 0.13 [0.06, 0.19], 0.16 [0.12, 0.20], and 0.20 [0.13, 0.27] in rare, occasional, and daily drinkers, respectively). In conclusion, the results of the present study indicated that salt-induced albuminuria was enhanced in subjects with higher drinking frequency, suggesting that salt restriction may have a stronger renoprotective effect in subjects with higher drinking frequency.
Collapse
Affiliation(s)
- Ryuichi Yoshimura
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan.,Division of Nephrology, Shimane University Hospital, Izumo, Japan
| | - Ryohei Yamamoto
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan. .,Health and Counseling Center, Osaka University, Toyonaka, Japan. .,Heatlth Promotion and Regulation, Department of Health Promotion Medicine, Osaka University Graduate School of Medicine, Toyonaka, Japan.
| | - Maki Shinzawa
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryohei Tomi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shingo Ozaki
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yoshiyuki Fujii
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takafumi Ito
- Division of Nephrology, Shimane University Hospital, Izumo, Japan
| | - Kazuaki Tanabe
- Department of Internal Medicine IV, Shimane University Faculty of Medicine, Izumo, Japan
| | | | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toshiki Moriyama
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan.,Health and Counseling Center, Osaka University, Toyonaka, Japan.,Heatlth Promotion and Regulation, Department of Health Promotion Medicine, Osaka University Graduate School of Medicine, Toyonaka, Japan
| |
Collapse
|
27
|
Sabbatini AR, Kararigas G. Estrogen-related mechanisms in sex differences of hypertension and target organ damage. Biol Sex Differ 2020; 11:31. [PMID: 32487164 PMCID: PMC7268741 DOI: 10.1186/s13293-020-00306-7] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Hypertension (HTN) is a primary risk factor for cardiovascular (CV) events, target organ damage (TOD), premature death and disability worldwide. The pathophysiology of HTN is complex and influenced by many factors including biological sex. Studies show that the prevalence of HTN is higher among adults aged 60 and over, highlighting the increase of HTN after menopause in women. Estrogen (E2) plays an important role in the development of systemic HTN and TOD, exerting several modulatory effects. The influence of E2 leads to alterations in mechanisms regulating the sympathetic nervous system, renin-angiotensin-aldosterone system, body mass, oxidative stress, endothelial function and salt sensitivity; all associated with a crucial inflammatory state and influenced by genetic factors, ultimately resulting in cardiac, vascular and renal damage in HTN. In the present article, we discuss the role of E2 in mechanisms accounting for the development of HTN and TOD in a sex-specific manner. The identification of targets with therapeutic potential would contribute to the development of more efficient treatments according to individual needs.
Collapse
Affiliation(s)
| | - Georgios Kararigas
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
28
|
Association of a Disrupted Dipping Pattern of Blood Pressure with Progression of Renal Injury during the Development of Salt-Dependent Hypertension in Rats. Int J Mol Sci 2020; 21:ijms21062248. [PMID: 32213948 PMCID: PMC7139748 DOI: 10.3390/ijms21062248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/19/2020] [Accepted: 03/21/2020] [Indexed: 12/28/2022] Open
Abstract
The aim of the present study is to investigate whether a disruption of the dipping pattern of blood pressure (BP) is associated with the progression of renal injury in Dahl salt-sensitive (DSS) hypertensive rats. Seven-week-old DSS rats were fed a high salt diet (HSD; 8% NaCl) for 10 weeks, followed by a transition to a normal salt diet (NSD; 0.3% NaCl) for 4 weeks. At baseline, NSD-fed DSS rats showed a dipper-type circadian rhythm of BP. By contrast, HSD for 5 days caused a significant increase in the difference between the active and inactive periods of BP with an extreme dipper type of BP, while proteinuria and renal tissue injury were not observed. Interestingly, HSD feeding for 10 weeks developed hypertension with a non-dipper pattern of BP, which was associated with obvious proteinuria and renal tissue injury. Four weeks after switching to an NSD, BP and proteinuria were significantly decreased, and the BP circadian rhythm returned to the normal dipper pattern. These data suggest that the non-dipper pattern of BP is associated with the progression of renal injury during the development of salt-dependent hypertension.
Collapse
|
29
|
|
30
|
Abstract
PURPOSE OF REVIEW This review will provide an in-depth coverage of the epidemiological and pre-clinical literature surrounding the role of dietary protein in hypertension, with a special emphasis on the history of our work on the Dahl salt-sensitive rat. RECENT FINDINGS Our studies have dedicated much effort into understanding the relationship between dietary protein and its effect on the development of salt-sensitive hypertension and renal injury. Our evidence over the last 15 years have demonstrated that both the source and amount of dietary protein can influence the severity of disease, where we have determined mechanisms related to immunity, the maternal environment during pregnancy, and more recently the gut microbiota, which significantly contribute to these diet-induced effects. Deeper understanding of these dietary protein-related mechanisms may provide insight on the plausibility of dietary modifications as future therapeutic avenues for hypertension and renal disease.
Collapse
|
31
|
Mattson DL. Immune mechanisms of salt-sensitive hypertension and renal end-organ damage. Nat Rev Nephrol 2019; 15:290-300. [PMID: 30804523 DOI: 10.1038/s41581-019-0121-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immune mechanisms have been recognized to have a role in the pathogenesis of hypertension, vascular disease and kidney damage in humans and animals for many decades. Contemporary advances in experimentation have permitted a deeper understanding of the mechanisms by which inflammation and immunity participate in cardiovascular disease, and multiple observations have demonstrated strong correlations between the discoveries made in animals and those made in patients with hypertension. Of note, striking phenotypic similarities have been observed in the infiltration of immune cells in the kidney and the development of end-organ damage in patients and animal models with sodium-sensitive hypertension. The available data suggest that an initial salt-induced increase in renal perfusion pressure, which is likely independent of immune mechanisms, induces the infiltration of immune cells into the kidney. The mechanisms mediating immune cell infiltration in the kidney are not well understood but likely involve tissue damage, the direct influence of salt to stimulate immune cell activation, sympathetic nerve stimulation or other factors. The infiltrating cells then release cytokines, free radicals and other factors that contribute to renal damage as well as increased retention of sodium and water and vascular resistance, which lead to the further development of hypertension.
Collapse
Affiliation(s)
- David L Mattson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
32
|
Abais-Battad JM, Alsheikh AJ, Pan X, Fehrenbach DJ, Dasinger JH, Lund H, Roberts ML, Kriegel AJ, Cowley AW, Kidambi S, Kotchen TA, Liu P, Liang M, Mattson DL. Dietary Effects on Dahl Salt-Sensitive Hypertension, Renal Damage, and the T Lymphocyte Transcriptome. Hypertension 2019; 74:854-863. [PMID: 31476910 DOI: 10.1161/hypertensionaha.119.12927] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Dahl salt-sensitive (SS) rat is an established model of SS hypertension and renal damage. In addition to salt, other dietary components were shown to be important determinants of hypertension in SS rats. With previous work eliminating the involvement of genetic differences, grain-fed SS rats from Charles River Laboratories (SS/CRL; 5L2F/5L79) were less susceptible to salt-induced hypertension and renal damage compared with purified diet-fed SS rats bred at the Medical College of Wisconsin (SS/MCW; 0.4% NaCl, AIN-76A). With the known role of immunity in hypertension, the present study characterized the immune cells infiltrating SS/MCW and SS/CRL kidneys via flow cytometry and RNA sequencing in T-cells isolated from the blood and kidneys of rats maintained on their respective parental diet or on 3 weeks of high salt (4.0% NaCl, AIN-76A). SS/CRL rats were protected from salt-induced hypertension (116.5±1.2 versus 141.9±14.4 mm Hg), albuminuria (21.7±3.5 versus 162.9±22.2 mg/d), and renal immune cell infiltration compared with SS/MCW. RNA-seq revealed >50% of all annotated genes in the entire transcriptome to be significantly differentially expressed in T-cells isolated from blood versus kidney, regardless of colony or chow. Pathway analysis of significantly differentially expressed genes between low and high salt conditions demonstrated changes related to inflammation in SS/MCW renal T-cells compared with metabolism-related pathways in SS/CRL renal T-cells. These functional and transcriptomic T-cell differences between SS/MCW and SS/CRL show that dietary components in addition to salt may influence immunity and the infiltration of immune cells into the kidney, ultimately impacting susceptibility to salt-induced hypertension and renal damage.
Collapse
Affiliation(s)
- Justine M Abais-Battad
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Ammar J Alsheikh
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Xiaoqing Pan
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Daniel J Fehrenbach
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - John Henry Dasinger
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Hayley Lund
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Michelle L Roberts
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Alison J Kriegel
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Allen W Cowley
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Srividya Kidambi
- Department of Medicine (S.K., T.A.K.), Medical College of Wisconsin
| | | | - Pengyuan Liu
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Mingyu Liang
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - David L Mattson
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| |
Collapse
|
33
|
Markakis K, Alam T, Jinadev P, Chapman A, Urwin A, Thabit H, Boulton AJM, Rutter MK, Leelarathna L. Continuous Subcutaneous Insulin Infusion Initiation Is Associated With Blood Pressure Reduction in Adults With Type 1 Diabetes. J Diabetes Sci Technol 2019; 13:691-697. [PMID: 30636544 PMCID: PMC6610598 DOI: 10.1177/1932296818822818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND In type 1 diabetes (T1D) continuous subcutaneous insulin infusion (CSII) use has been associated with lower risks for mortality and diabetes-related complications when compared to multiple daily injections. There are limited data on the effect of CSII on blood pressure (BP). We aimed to evaluate the relationship between CSII initiation and BP changes. METHODS We performed a retrospective, single-center, observational study including all adults with T1D who initiated CSII between 2001 and 2014. Mean systolic BP levels were calculated from values obtained up to 15 months before and up 15 months after CSII initiation. RESULTS We studied 436 adults (46% male; mean [95% CI] age: 45 [36, 54] years, duration of diabetes: 20.3 [19.1, 21.5] years). CSII was associated with the following changes (post minus pre CSII levels) in mean (95% CI) SBP and DBP levels, respectively: whole cohort (N = 436): -2 (-1, -3), P < .001 and -2 (-1, -2), P < .001; those taking stable antihypertensive medication (n = 118): -4 (-6, -2), P < .001 and -2 (-3, -1), P < .001; antihypertensive treatment-naïve patients (n = 279): -1 (-2, 0), P = .147 and -1 (-2, 0), P = .002. In multivariate regression analysis including age, sex, diabetes duration, pre-CSII BP, HbA1c and weight changes, only pre-CSII BP showed an independent association with BP drop in all patients' groups. Once adjusted for pre-CSII BP there was no significant difference between those on antihypertensive drugs and antihypertensive treatment-naïve individuals. CONCLUSIONS We showed BP reduction in association with CSII therapy particularly in those on antihypertensive medication. These original findings may partly explain the cardiovascular and mortality benefits associated with CSII therapy.
Collapse
Affiliation(s)
- Konstantinos Markakis
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
| | - Tayiba Alam
- School of Medical Sciences, University
of Manchester, Manchester, UK
| | - Pushpa Jinadev
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
| | - Alyson Chapman
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
| | - Andrea Urwin
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
| | - Hood Thabit
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
- School of Medical Sciences, University
of Manchester, Manchester, UK
| | - Andrew J. M. Boulton
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
- Division of Diabetes, Endocrinology and
Gastroenterology, University of Manchester, Manchester, UK
| | - Martin K. Rutter
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
- Division of Diabetes, Endocrinology and
Gastroenterology, University of Manchester, Manchester, UK
| | - Lalantha Leelarathna
- Manchester Diabetes Centre, Manchester
University NHS Foundation Trust, Manchester Academic Health Science Centre,
Manchester, UK
- Division of Diabetes, Endocrinology and
Gastroenterology, University of Manchester, Manchester, UK
- Lalantha Leelarathna, PhD, Manchester
Diabetes Centre, Hathersage Rd, Manchester, M13 9WL, UK.
| |
Collapse
|
34
|
Fehrenbach DJ, Abais-Battad JM, Dasinger JH, Lund H, Mattson DL. Salt-sensitive increase in macrophages in the kidneys of Dahl SS rats. Am J Physiol Renal Physiol 2019; 317:F361-F374. [PMID: 31215801 DOI: 10.1152/ajprenal.00096.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Studies of Dahl salt-sensitive (SS) rats have shown that renal CD3+ T cells and ED-1+ macrophages are involved in the development of salt-sensitive hypertension and renal damage. The present study demonstrated that the increase in renal immune cells, which accompanies renal hypertrophy and albuminuria in high-salt diet-fed Dahl SS rats, is absent in Sprague-Dawley and SSBN13 rats that are protected from the SS disease phenotype. Flow cytometric analysis demonstrated that >70% of the immune cells in the SS kidney are M1 macrophages. PCR profiling of renal myeloid cells showed a salt-induced upregulation in 9 of 84 genes related to Toll-like receptor signaling, with notable upregulation of the Toll-like receptor 4/CD14/MD2 complex. Because of the prominent increase in macrophages in the SS kidney, we used liposome-encapsulated clodronate (Clod) to deplete macrophages and assess their contribution to salt-sensitive hypertension and renal damage. Dahl SS animals were administered either Clod-containing liposomes (Clod-Lipo), Clod, or PBS-containing liposomes as a vehicle control. Clod-Lipo treatment depleted circulating and splenic macrophages by ∼50%; however, contrary to our hypothesis, Clod-Lipo-treated animals developed an exacerbated salt-sensitive response with respect to blood pressure and albuminuria, which was accompanied by increased renal T and B cells. Interestingly, those treated with Clod also demonstrated an exacerbated phenotype, but it was less severe than Clod-Lipo-treated animals and independent of changes to the number of renal immune cells. Here, we have shown that renal macrophages in Dahl SS animals sustain a M1 proinflammatory phenotype in response to increased dietary salt and highlighted potential adverse effects of Clod-Lipo macrophage depletion.
Collapse
Affiliation(s)
- Daniel J Fehrenbach
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | | | - John Henry Dasinger
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - Hayley Lund
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin, Wauwatosa, Wisconsin
| |
Collapse
|
35
|
Krajcoviechova A, Marois-Blanchet FC, Troyanov S, Harvey F, Dumas P, Tremblay J, Cifkova R, Awadalla P, Madore F, Hamet P. Uromodulin in a Pathway Between Decreased Renal Urate Excretion and Albuminuria. Am J Hypertens 2019; 32:384-392. [PMID: 30551216 DOI: 10.1093/ajh/hpy190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 11/25/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The mechanism explaining the inverse association between renal urate and albumin excretion remains unclear. First, we evaluated the impact of candidate variants in the main urate transporter genes (i.e., SLC2A9, SLC22A12, ABCG2) on the association between fractional excretion of uric acid (FEUA) and urinary albumin/creatinine ratio (uACR). Second, we examined uromodulin and sodium excretion as mediators of the association between FEUA and uACR. METHODS We performed cross-sectional analysis of 737 French Canadians from the CARTaGENE cohort, a random sample of the Quebec population aged 40-69 years (a total of 20,004 individuals). Individuals with available genotyping and urinary data were obtained from a sub-study including gender-matched pairs with high and low Framingham Risk Score and vascular rigidity index. We further excluded individuals with an estimated glomerular filtration rate <60 ml/min/1.73 m2, glycosuria, and use of confounding medication. A spot urine sample was analyzed. Genotyping was performed using the Illumina Omni2.5-8 BeadChips. Genetic variants were analyzed using an additive model. RESULTS Final analyses included 593 individuals (45.5% of men; mean age 54.3 ± 8.6). We observed an antagonistic interaction between rs13129697 variant of the SLC2A9 gene and FEUA tertiles on uACR (P = 0.002). Using the mediation analysis, uromodulin explained 32%, fractional excretion of sodium (FENa) 44%, and uromodulin together with FENa explained 70% of the inverse relationship between FEUA and uACR. Bootstrapping process confirmed the role of both mediators. CONCLUSIONS Our data suggest that the association of albuminuria with decreased renal urate excretion may be modified by the transporter SLC2A9, and mediated by uromodulin and sodium handling.
Collapse
Affiliation(s)
- Alena Krajcoviechova
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
- Centre de recherche du CHUM, Montréal, Quebec, Canada
| | | | - Stephan Troyanov
- Division of Nephrology, Department of Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Quebec, Canada
| | | | - Pierre Dumas
- Centre de recherche du CHUM, Montréal, Quebec, Canada
| | | | - Renata Cifkova
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
| | - Philip Awadalla
- Centre hospitalier universitaire Sainte-Justine, CHU Sainte-Justine, Montréal, Quebec, Canada
| | - Francois Madore
- Division of Nephrology, Department of Medicine, Hôpital du Sacré-Coeur de Montréal, Montréal, Quebec, Canada
| | - Pavel Hamet
- Centre de recherche du CHUM, Montréal, Quebec, Canada
| |
Collapse
|
36
|
Kurtz TW, DiCarlo SE, Pravenec M, Morris RC. Changing views on the common physiologic abnormality that mediates salt sensitivity and initiation of salt-induced hypertension: Japanese research underpinning the vasodysfunction theory of salt sensitivity. Hypertens Res 2018; 42:6-18. [DOI: 10.1038/s41440-018-0122-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/24/2022]
|
37
|
Abais-Battad JM, Mattson DL. Influence of dietary protein on Dahl salt-sensitive hypertension: a potential role for gut microbiota. Am J Physiol Regul Integr Comp Physiol 2018; 315:R907-R914. [PMID: 30133303 PMCID: PMC6295491 DOI: 10.1152/ajpregu.00399.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 07/30/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
High blood pressure affects 1.39 billion adults across the globe and is the leading preventable cause of death worldwide. Hypertension is a multifaceted disease with known genetic and environmental factors contributing to its progression. Our studies utilizing the Dahl salt-sensitive (SS) rat have demonstrated the remarkable influence of dietary protein and maternal environment on the development of hypertension and renal damage in response to high salt. There is growing interest in the relationship between the microbiome and hypertension, with gut dysbiosis being correlated to a number of pathologies. This review summarizes the current literature regarding the interplay among dietary protein, the gut microbiota, and hypertension. These studies may provide insight into the effects we have observed between diet and hypertension in Dahl SS rats and, we hope, lead to new perspectives where potential dietary interventions or microbiota manipulations could serve as plausible therapies for hypertension.
Collapse
Affiliation(s)
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
38
|
The pivotal role of renal vasodysfunction in salt sensitivity and the initiation of salt-induced hypertension. Curr Opin Nephrol Hypertens 2018; 27:83-92. [DOI: 10.1097/mnh.0000000000000394] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
39
|
Mishra S, Ingole S, Jain R. Salt sensitivity and its implication in clinical practice. Indian Heart J 2017; 70:556-564. [PMID: 30170653 PMCID: PMC6116721 DOI: 10.1016/j.ihj.2017.10.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/01/2017] [Accepted: 10/10/2017] [Indexed: 02/06/2023] Open
Abstract
Hypertension (HTN) is a complex multi-factorial disease and is considered one of the foremost modifiable risk factors for stroke, heart failure, ischemic heart disease and renal dysfunction. Over the past century, salt and its linkage to HTN and cardiovascular (CV) mortality has been the subject of intense scientific scrutiny. There is now consensus that different individuals have different susceptibilities to blood pressure (BP)-raising effects of salt and this susceptiveness is called as salt sensitivity. Several renal and extra-renal mechanisms are believed to play a role. Blunted activity of the renin–angiotensin–aldosterone system (RAAS), adrenal Rac1-MR-Sgk1-NCC/ENaC pathway, renal SNS-GR-WNK4-NCC pathway, defect of membrane ion transportation, inflammation and abnormalities of Na+/Ca2+ exchange have all been implicated as pathophysiological basis for salt sensitive HTN. While salt restriction is definitely beneficial recent observation suggests that treatment with Azilsartan may improve salt sensitivity by selectively reducing renal proximal tubule Na+/H+ exchange. This encourages the future potential benefits of recognizing and therapeutically addressing the salt sensitive phenotype in humans.
Collapse
|
40
|
Significant natriuretic and antihypertensive action of the epithelial sodium channel blocker amiloride in diabetic patients with and without nephropathy. J Hypertens 2017; 34:1621-9. [PMID: 27214087 DOI: 10.1097/hjh.0000000000000967] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Diabetic nephropathy is associated with aberrant glomerular filtration of serine proteases. The study was designed to test the hypothesis that the epithelial sodium channel is activated proteolytically by urine plasmin in diabetic nephropathy and mediates renal sodium retention. METHODS In an open-label intervention study on type 1 diabetes patients on standardized NaCl intake (200 mmol/day) with (n = 15) and without diabetic nephropathy (control, n = 12), urinary Na excretion in response to oral amiloride (20 or 40 mg/day for 2 days) was compared. RESULTS A total of 27 patients completed the study and nine diabetic nephropathy and eight control study participants were compliant (24-h urine Na excretion of 200 mmol ± 30%). Amiloride increased significantly total and fractional Na excretion in both groups. Total natriuresis and weight loss were significantly larger in the control group compared with diabetic nephropathy at day 1 of amiloride, whereas fractional Na excretion did not differ. Amiloride intervention increased plasma renin concentration only in diabetic nephropathy group; it reduced SBP in both groups, whereas DBP was reduced in diabetic nephropathy group only. Albuminuria was reduced significantly by amiloride in diabetic nephropathy group. Urine total amiloride concentration was not different between groups (12 ± 1 and 16 ± 1 μmol/l, respectively). Urine total plasminogen and active plasmin were reduced after amiloride in diabetic nephropathy. CONCLUSION Amiloride increased renal Na excretion, reduced blood pressure, albuminuria, and total and active plasmin in urine. It is concluded that epithelial sodium channel is an attractive target to attain blood pressure control in long-term type I diabetes with no enhanced activity associated with nephropathy.
Collapse
|
41
|
Naser AM, Unicomb L, Doza S, Ahmed KM, Rahman M, Uddin MN, Quraishi SB, Selim S, Shamsudduha M, Burgess W, Chang HH, Gribble MO, Clasen TF, Luby SP. Stepped-wedge cluster-randomised controlled trial to assess the cardiovascular health effects of a managed aquifer recharge initiative to reduce drinking water salinity in southwest coastal Bangladesh: study design and rationale. BMJ Open 2017; 7:e015205. [PMID: 28864689 PMCID: PMC5588995 DOI: 10.1136/bmjopen-2016-015205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 06/16/2017] [Accepted: 07/13/2017] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION Saltwater intrusion and salinisation have contributed to drinking water scarcity in many coastal regions globally, leading to dependence on alternative sources for water supply. In southwest coastal Bangladesh, communities have few options but to drink brackish groundwater which has been associated with high blood pressure among the adult population, and pre-eclampsia and gestational hypertension among pregnant women. Managed aquifer recharge (MAR), the purposeful recharge of surface water or rainwater to aquifers to bring hydrological equilibrium, is a potential solution for salinity problem in southwest coastal Bangladesh by creating a freshwater lens within the brackish aquifer. Our study aims to evaluate whether consumption of MAR water improves human health, particularly by reducing blood pressure among communities in coastal Bangladesh. METHODS AND ANALYSIS The study employs a stepped-wedge cluster-randomised controlled community trial design in 16 communities over five monthly visits. During each visit, we will collect data on participants' source of drinking and cooking water and measure the salinity level and electrical conductivity of household stored water. At each visit, we will also measure the blood pressure of participants ≥20 years of age and pregnant women and collect urine samples for urinary sodium and protein measurements. We will use generalised linear mixed models to determine the association of access to MAR water on blood pressure of the participants. ETHICS AND DISSEMINATION The study protocol has been reviewed and approved by the Institutional Review Boards of the International Centre for Diarrheal Disease Research, Bangladesh (icddr,b). Informed written consent will be taken from all the participants. This study is funded by Wellcome Trust, UK. The study findings will be disseminated to the government partners, at research conferences and in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT02746003; Pre-results.
Collapse
Affiliation(s)
- Abu Mohd Naser
- Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Leanne Unicomb
- Environmental Health & Interventions Unit, Enteric and Respiratory Infections Program, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Solaiman Doza
- Environmental Health & Interventions Unit, Enteric and Respiratory Infections Program, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | | | - Mahbubur Rahman
- Environmental Health & Interventions Unit, Enteric and Respiratory Infections Program, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Mohammad Nasir Uddin
- Environmental Health & Interventions Unit, Enteric and Respiratory Infections Program, Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Shamshad B Quraishi
- Analytical Chemistry Laboratory, Atomic Energy Centre, Bangladesh Atomic EnergyCommission, Dhaka, Bangladesh
| | - Shahjada Selim
- Department of Endocrinology & Metabolism, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Mohammad Shamsudduha
- Institute for Risk and Disaster Reduction, Departmentof Geography, University College London, London, UK
| | - William Burgess
- Department of Earth Sciences, University College London, London, UK
| | - Howard H Chang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Matthew O Gribble
- Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Thomas F Clasen
- Department of Environmental Health Sciences, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Stephen P Luby
- Stanford Woods Institute for the Environment & Freeman Spogli Institute for International Studies, Stanford University, Stanford, California, USA
| |
Collapse
|
42
|
Wang H, Romero CA, Masjoan Juncos JX, Monu SR, Peterson EL, Carretero OA. Effect of salt intake on afferent arteriolar dilatation: role of connecting tubule glomerular feedback (CTGF). Am J Physiol Renal Physiol 2017; 313:F1209-F1215. [PMID: 28835421 DOI: 10.1152/ajprenal.00320.2017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/10/2017] [Accepted: 08/16/2017] [Indexed: 01/13/2023] Open
Abstract
Afferent arteriole (Af-Art) resistance is modulated by two intrinsic nephron feedbacks: 1) the vasoconstrictor tubuloglomerular feedback (TGF) mediated by Na+-K+-2Cl- cotransporters (NKCC2) in the macula densa and blocked by furosemide and 2) the vasodilator connecting tubule glomerular feedback (CTGF), mediated by epithelial Na+ channels (ENaC) in the connecting tubule and blocked by benzamil. High salt intake reduces Af-Art vasoconstrictor ability in Dahl salt-sensitive rats (Dahl SS). Previously, we measured CTGF indirectly, by differences between TGF responses with and without CTGF inhibition. We recently developed a new method to measure CTGF more directly by simultaneously inhibiting NKCC2 and the Na+/H+ exchanger (NHE). We hypothesize that in vivo during simultaneous inhibition of NKCC2 and NHE, CTGF causes an Af-Art dilatation revealed by an increase in stop-flow pressure (PSF) in Dahl SS and that is enhanced with a high salt intake. In the presence of furosemide alone, increasing nephron perfusion did not change the PSF in either Dahl salt-resistant rats (Dahl SR) or Dahl SS. When furosemide and an NHE inhibitor, dimethylamiloride, were perfused simultaneously, an increase in tubular flow caused Af-Art dilatation that was demonstrated by an increase in PSF. This increase was greater in Dahl SS [4.5 ± 0.4 (SE) mmHg] than in Dahl SR (2.5 ± 0.3 mmHg; P < 0.01). We confirmed that CTGF causes this vasodilation, since benzamil completely blocked this effect. However, a high salt intake did not augment the Af-Art dilatation. We conclude that during simultaneous inhibition of NKCC2 and NHE in the nephron, CTGF induces Af-Art dilatation and a high salt intake failed to enhance this effect.
Collapse
Affiliation(s)
- Hong Wang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Cesar A Romero
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - J X Masjoan Juncos
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan; and
| | - Sumit R Monu
- 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
|
43
|
Kurtz TW, DiCarlo SE, Pravenec M, Morris RC. An Appraisal of Methods Recently Recommended for Testing Salt Sensitivity of Blood Pressure. J Am Heart Assoc 2017; 6:JAHA.117.005653. [PMID: 28365569 PMCID: PMC5533040 DOI: 10.1161/jaha.117.005653] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Theodore W Kurtz
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA
| | | | - Michal Pravenec
- Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - R Curtis Morris
- Department of Medicine, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
44
|
Clemmer JS, Pruett WA, Coleman TG, Hall JE, Hester RL. Mechanisms of blood pressure salt sensitivity: new insights from mathematical modeling. Am J Physiol Regul Integr Comp Physiol 2016; 312:R451-R466. [PMID: 27974315 DOI: 10.1152/ajpregu.00353.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 12/06/2016] [Accepted: 12/06/2016] [Indexed: 12/24/2022]
Abstract
Mathematical modeling is an important tool for understanding quantitative relationships among components of complex physiological systems and for testing competing hypotheses. We used HumMod, a large physiological model, to test hypotheses of blood pressure (BP) salt sensitivity. Systemic hemodynamics, renal, and neurohormonal responses to chronic changes in salt intake were examined during normal renal function, fixed low or high plasma angiotensin II (ANG II) levels, bilateral renal artery stenosis, increased renal sympathetic nerve activity (RSNA), and decreased nephron numbers. Simulations were run for 4 wk at salt intakes ranging from 30 to 1,000 mmol/day. Reducing functional kidney mass or fixing ANG II increased salt sensitivity. Salt sensitivity, associated with inability of ANG II to respond to changes in salt intake, occurred with smaller changes in renal blood flow but greater changes in glomerular filtration rate, renal sodium reabsorption, and total peripheral resistance (TPR). However, clamping TPR at normal or high levels had no major effect on salt sensitivity. There were no clear relationships between BP salt sensitivity and renal vascular resistance or extracellular fluid volume. Our robust mathematical model of cardiovascular, renal, endocrine, and sympathetic nervous system physiology supports the hypothesis that specific types of kidney dysfunction, associated with impaired regulation of ANG II or increased tubular sodium reabsorption, contribute to BP salt sensitivity. However, increased preglomerular resistance, increased RSNA, or inability to decrease TPR does not appear to influence salt sensitivity. This model provides a platform for testing competing concepts of long-term BP control during changes in salt intake.
Collapse
Affiliation(s)
- John S Clemmer
- Department of Physiology and Biophysics, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - W Andrew Pruett
- Department of Physiology and Biophysics, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Thomas G Coleman
- Department of Physiology and Biophysics, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - John E Hall
- Department of Physiology and Biophysics, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Robert L Hester
- Department of Physiology and Biophysics, Center for Computational Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
45
|
Mahajan A, Rodan AR, Le TH, Gaulton KJ, Haessler J, Stilp AM, Kamatani Y, Zhu G, Sofer T, Puri S, Schellinger JN, Chu PL, Cechova S, van Zuydam N, Arnlov J, Flessner MF, Giedraitis V, Heath AC, Kubo M, Larsson A, Lindgren CM, Madden PAF, Montgomery GW, Papanicolaou GJ, Reiner AP, Sundström J, Thornton TA, Lind L, Ingelsson E, Cai J, Martin NG, Kooperberg C, Matsuda K, Whitfield JB, Okada Y, Laurie CC, Morris AP, Franceschini N. Trans-ethnic Fine Mapping Highlights Kidney-Function Genes Linked to Salt Sensitivity. Am J Hum Genet 2016; 99:636-646. [PMID: 27588450 PMCID: PMC5011075 DOI: 10.1016/j.ajhg.2016.07.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/08/2016] [Indexed: 01/09/2023] Open
Abstract
We analyzed genome-wide association studies (GWASs), including data from 71,638 individuals from four ancestries, for estimated glomerular filtration rate (eGFR), a measure of kidney function used to define chronic kidney disease (CKD). We identified 20 loci attaining genome-wide-significant evidence of association (p < 5 × 10(-8)) with kidney function and highlighted that allelic effects on eGFR at lead SNPs are homogeneous across ancestries. We leveraged differences in the pattern of linkage disequilibrium between diverse populations to fine-map the 20 loci through construction of "credible sets" of variants driving eGFR association signals. Credible variants at the 20 eGFR loci were enriched for DNase I hypersensitivity sites (DHSs) in human kidney cells. DHS credible variants were expression quantitative trait loci for NFATC1 and RGS14 (at the SLC34A1 locus) in multiple tissues. Loss-of-function mutations in ancestral orthologs of both genes in Drosophila melanogaster were associated with altered sensitivity to salt stress. Renal mRNA expression of Nfatc1 and Rgs14 in a salt-sensitive mouse model was also reduced after exposure to a high-salt diet or induced CKD. Our study (1) demonstrates the utility of trans-ethnic fine mapping through integration of GWASs involving diverse populations with genomic annotation from relevant tissues to define molecular mechanisms by which association signals exert their effect and (2) suggests that salt sensitivity might be an important marker for biological processes that affect kidney function and CKD in humans.
Collapse
Affiliation(s)
- Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Aylin R Rodan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75229, USA
| | - Thu H Le
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Kyle J Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Adrienne M Stilp
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Gu Zhu
- Genetic Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Tamar Sofer
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Sanjana Puri
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75229, USA
| | - Jeffrey N Schellinger
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75229, USA
| | - Pei-Lun Chu
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Sylvia Cechova
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Natalie van Zuydam
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Johan Arnlov
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala 751 85, Sweden; School of Health and Social Studies, Dalarna University, Falun 791 88, Sweden
| | - Michael F Flessner
- National Institute of Diabetes, Digestive, and Kidney Disease, NIH, Bethesda, MD 20892, USA
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala 752 37, Sweden
| | - Andrew C Heath
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michiaki Kubo
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Anders Larsson
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala 751 85, Sweden
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7BN, UK
| | - Pamela A F Madden
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Grant W Montgomery
- Molecular Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - George J Papanicolaou
- Epidemiology Branch, Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Alex P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Johan Sundström
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala 751 85, Sweden
| | - Timothy A Thornton
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala 751 85, Sweden
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala 752 37, Sweden; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jianwen Cai
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nicholas G Martin
- Genetic Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Koichi Matsuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - John B Whitfield
- Genetic Epidemiology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan; Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Cathy C Laurie
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK.
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27514, USA.
| |
Collapse
|
46
|
Elijovich F, Weinberger MH, Anderson CAM, Appel LJ, Bursztyn M, Cook NR, Dart RA, Newton-Cheh CH, Sacks FM, Laffer CL. Salt Sensitivity of Blood Pressure: A Scientific Statement From the American Heart Association. Hypertension 2016; 68:e7-e46. [PMID: 27443572 DOI: 10.1161/hyp.0000000000000047] [Citation(s) in RCA: 361] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
47
|
Fellner RC, Guan Z, Cook AK, Pollock DM, Inscho EW. Endothelin contributes to blunted renal autoregulation observed with a high-salt diet. Am J Physiol Renal Physiol 2015; 309:F687-96. [PMID: 26246513 DOI: 10.1152/ajprenal.00641.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 07/29/2015] [Indexed: 01/03/2023] Open
Abstract
Autoregulation of renal blood flow (RBF) is an essential function of the renal microcirculation that has been previously shown to be blunted by excessive dietary salt. Endogenous endothelin 1 (ET-1) is increased following a high-salt (HS) diet and contributes to the control of RBF but the differential effects of ET-1 on renal microvessel autoregulation in response to HS remain to be established. We hypothesized that a HS diet increases endothelin receptor activation in normal Sprague-Dawley rats and blunts autoregulation of RBF. The role of ET-1 in the blunted autoregulation produced by a HS diet was assessed in vitro and in vivo using the blood-perfused juxtamedullary nephron preparation and anesthetized rats, respectively. Using highly selective antagonists, we observed that blockade of either ETA or ETB receptors was sufficient to restore normal autoregulatory behavior in afferent arterioles from HS-fed rats. Additionally, normal autoregulatory behavior was restored in vivo in HS-fed rats by simultaneous ETA and ETB receptor blockade, whereas blockade of ETB receptors alone showed significant improvement of normal autoregulation of RBF. Consistent with this observation, autoregulation of RBF in ETB receptor-deficient rats fed HS was similar to both ETB-deficient rats and transgenic control rats on normal-salt diets. These data support the hypothesis that endogenous ET-1, working through ETB and possibly ETA receptors, contributes to the blunted renal autoregulatory behavior in rats fed a HS diet.
Collapse
Affiliation(s)
- Robert C Fellner
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia
| | - Zhengrong Guan
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anthony K Cook
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - David M Pollock
- Section of Experimental Medicine, Department of Medicine, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; and Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Edward W Inscho
- Department of Physiology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia; Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
48
|
Plasmin in urine from patients with type 2 diabetes and treatment-resistant hypertension activates ENaC in vitro. J Hypertens 2015; 32:1672-7; discussion 1677. [PMID: 24805959 DOI: 10.1097/hjh.0000000000000216] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Aberrant filtration of plasminogen from plasma and subsequent activation to plasmin in the urinary space may activate proteolytically the epithelial sodium channel, ENaC. In conditions with chronic albuminuria, this may cause hypertension. It was hypothesized that patients with type 2 diabetes mellitus (T2DM) and treatment-resistant hypertension excrete plasmin(ogen) in urine in proportion to albumin and that plasmin confers to urine the ability to activate ENaC. METHOD Patients (n = 113) with T2DM and resistant hypertension, defined as systolic blood pressure (SBP) more than 130 mmHg and/or diastolic blood pressure (DBP) more than 80 mmHg despite use of at least three drugs with one diuretic and one renin-angiotensin system inhibitor, were included. Urine was analyzed for albumin, creatinine, plasmin(ogen), protease activity, and ability to activate inward current in single collecting duct cells. RESULTS Mean ambulatory SBP/DBP was 143 ± 1/77 ± 0.7 mmHg; HbA1c 7.35%; and eGFR 81.0 ml/min per 1.73 m (geometric means). Patients with microalbuminuria (39%) and macroalbuminuria (13%) displayed significantly elevated levels of urinary plasmin(ogen) normalized to urine creatinine compared with patients with normal excretion of albumin (48%). Urinary plasminogen correlated significantly to urine albumin. Western immunoblotting and gelatine zymography confirmed active plasmin in urine samples from patients with microalbuminuria and macroalbuminuria. Single collecting duct cells displayed significantly increased, amiloride-sensitive, inward current when superfused with urine from albuminuric patients compared with patients with normal albumin excretion. Urinary plasminogen/creatinine ratio correlated significantly with 24-h ambulatory blood pressure. CONCLUSION Aberrant presence of plasmin in preurine may inappropriately activate ENaC in patients with type 2 diabetes and microalbuminuria. This may contribute to treatment-resistant hypertension.
Collapse
|
49
|
Cao W, Li A, Wang L, Zhou Z, Su Z, Bin W, Wilcox CS, Hou FF. A Salt-Induced Reno-Cerebral Reflex Activates Renin-Angiotensin Systems and Promotes CKD Progression. J Am Soc Nephrol 2015; 26:1619-33. [PMID: 25635129 DOI: 10.1681/asn.2014050518] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 09/10/2014] [Indexed: 12/30/2022] Open
Abstract
Salt intake promotes progression of CKD by uncertain mechanisms. We hypothesized that a salt-induced reno-cerebral reflex activates a renin-angiotensin axis to promote CKD. Sham-operated and 5/6-nephrectomized rats received a normal-salt (0.4%), low-salt (0.02%), or high-salt (4%) diet for 2 weeks. High salt in 5/6-nephrectomized rats increased renal NADPH oxidase, inflammation, BP, and albuminuria. Furthermore, high salt activated the intrarenal and cerebral, but not the systemic, renin-angiotensin axes and increased the activity of renal sympathetic nerves and neurons in the forebrain of these rats. Renal fibrosis was increased 2.2-fold by high versus low salt, but intracerebroventricular tempol, losartan, or clonidine reduced this fibrosis by 65%, 69%, or 59%, respectively, and renal denervation or deafferentation reduced this fibrosis by 43% or 38%, respectively (all P<0.05). Salt-induced fibrosis persisted after normalization of BP with hydralazine. These data suggest that the renal and cerebral renin-angiotensin axes are interlinked by a reno-cerebral reflex that is activated by salt and promotes oxidative stress, fibrosis, and progression of CKD independent of BP.
Collapse
Affiliation(s)
- Wei Cao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; and
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; and
| | - Liangliang Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; and
| | - Zhanmei Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; and
| | - Zhengxiu Su
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; and
| | - Wei Bin
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; and
| | - Christopher S Wilcox
- Center for Hypertension, Kidney, and Vascular Research, Georgetown University, Washington, DC
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China; and
| |
Collapse
|
50
|
Andersen H, Friis UG, Hansen PBL, Svenningsen P, Henriksen JE, Jensen BL. Diabetic nephropathy is associated with increased urine excretion of proteases plasmin, prostasin and urokinase and activation of amiloride-sensitive current in collecting duct cells. Nephrol Dial Transplant 2015; 30:781-9. [PMID: 25609736 DOI: 10.1093/ndt/gfu402] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/02/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is associated with hypertension, expanded extracellular volume and impaired renal Na(+) excretion. It was hypothesized that aberrant glomerular filtration of serine proteases in DN causes proteolytic activation of the epithelial sodium channel (ENaC) in the kidney by excision of an inhibitory peptide tract from the γ subunit. METHODS In a cross-sectional design, urine, plasma and clinical data were collected from type 1 diabetic patients with DN (n = 19) and matched normoalbuminuric type 1 diabetics (controls, n = 20). Urine was examined for proteases by western immunoblotting, patch clamp and ELISA. Urine exosomes were isolated to elucidate potential cleavage of γENaC by a monoclonal antibody directed against the 'inhibitory' peptide tract. RESULTS Compared with control, DN patients displayed significantly higher blood pressure and urinary excretion of plasmin(ogen), prostasin and urokinase that correlated directly with urine albumin. Western blotting confirmed plasmin, prostasin and urokinase in urine from the DN group predominantly. Urine from DN evoked a significantly larger amiloride-sensitive inward current in single collecting duct cells compared with controls. Immunoblotting of urine exosomes showed aquaporin 2 in all patient samples. Exosomes displayed a virtual absence of intact γENaC while moieties compatible with cleavage by furin only, were shown in both groups. Proteolytic cleavage by the extracellular serine proteases plasmin or prostasin was observed in DN samples predominantly. CONCLUSION DN is associated with increased urinary excretion of plasmin, prostasin and urokinase and proteolytic activation of ENaC that might contribute to impaired renal Na(+) excretion and hypertension.
Collapse
Affiliation(s)
- Henrik Andersen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark Department of Endocrinology, Odense University Hospital, Odense C, Denmark
| | - Ulla G Friis
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Pernille B L Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Per Svenningsen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Jan Erik Henriksen
- Department of Endocrinology, Odense University Hospital, Odense C, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|