1
|
Bansal S, Puzantian H, Townsend RR. Rising Prevalence of Obesity and Primary Hyperaldosteronism: Co-incidence or Connected Circumstances Leading to Hypertension? A Narrative Review. J Gen Intern Med 2024:10.1007/s11606-024-09081-2. [PMID: 39414738 DOI: 10.1007/s11606-024-09081-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/24/2024] [Indexed: 10/18/2024]
Abstract
While obesity and its associated complications, mainly diabetes and hypertension, have been the largest public health problems of modern world, the emerging data suggests an increasing prevalence of primary hyperaldosteronism (PA) as one of the most common undiagnosed causes of hypertension. We believe that rising prevalence of PA in the era of high rates of obesity is likely not a chance finding but is deeply intersected with the rising rates of obesity. Higher serum aldosterone concentrations and urinary aldosterone excretion have been observed in patients with increased body mass index or larger waist circumference. The in vitro and pre-clinical studies suggest that adipocytes not only synthesize and secrete aldosterone but also release factors which stimulate production of aldosterone from adrenal glands. Aldosterone excess causing ligand-dependent activation of the mineralocorticoid receptor (MR) has increasingly been recognized as one of the important mechanisms of obesity-related hypertension. The aldosterone excess in these cases can be labelled as acquired hyperaldosteronism to differentiate it from the non-obesity related classical cases of PA. Because of serious consequences, recognizing aldosterone excess in obesity is important, as it gives a more compelling reason for weight loss and guidance to choosing pharmacotherapy wisely. Dietary sodium restriction and mineralocorticoid receptor antagonists play important roles in the management of PA associated with obesity.
Collapse
Affiliation(s)
- Shweta Bansal
- Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, 7703 Floyd Curl Drive, MSC 7882, San Antonio, TX, 78229, USA.
| | - Houry Puzantian
- Hariri School of Nursing, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Raymond R Townsend
- Division of Renal Electrolyte and Hypertension, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
2
|
Zhou W, Xu C, Niu J, Xiong Y, He Z, Xu H, Zhang M, Wang H, Xu Q, Wang X, Wang Z. Inhibitory effects of Eplerenone on angiogenesis via modulating SGK1/TGF-β pathway in contralateral kidney of CKD pregnancy rats. Cell Signal 2024; 122:111346. [PMID: 39147296 DOI: 10.1016/j.cellsig.2024.111346] [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: 03/12/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Eplerenone is a selective aldosterone receptor blocker that is effective in preventing the progression of chroinic kidney disease (CKD). However, its mechanism and role in CKD pregnancy still remain uncertain. The aim of this study was to evaluate whether eplerenone could attenuated the fibrosis of unilateral ureteral obstruction (UUO) pregnant rats' contralateral kidney, improved pregnancy outcome and explore its therapeutic mechanisms. METHODS A pregnancy rat model of UUO established, female Wistar rats were randomly assigned into sham-operated group (Sham group),sham-operated combined pregnancy group (SP group), unilateral ureteral obstruction combined pregnancy group (UUO + Pregnancy group), unilateral ureteral obstruction combined pregnancy, administered eplerenone (UUO + Pregnancy+Eplerenone group). On the 18th day of pregnancy, the rats were placed in a metabolic cage, 24 h urine was collected and stored at -80 °C. Next day, all animals were euthanized, and serum was collected by centrifugation and stored at -20 °C. Then the right kidney was extracted, a part of the kidney was placed in 4% paraformaldehyde for morphology, immunohistochemical staining, and immunofluorescence staining, and the other part was placed in a - 80 °C refrigerator for RNA and protein extraction. In vitro, HUVECs was treated with aldosterone, progesterone and estradiol, VEGFA and its receptor blocker bevacizumab. The ability of proliferation, migration and tubularization of HUVECs was detected by CCK-8, scratch wound assay and endothelial tube formation assay. And the co-expression of CD34 and α-SMA of HUVECs was detected by Flow cytometry. RESULTS Immunofluorescence results showed that the co-expression of CD34 and α-SMA increased in the UUO + Pregnancy group was significantly increased. The expression of SGK-1, TGFβ-1, Smad2, Smad3, VEGF-A, VEGFR2, CD34, α-SMA and Collagen I was significantly higher in the kidneys of the UUO + Pregnancy group compared to the Sham group and SP group. Eplerenone inhibited the expression of those results. In vitro, the ability of proliferation, migration and tubularization was increased after treated with aldosterone, aldosterone with progesterone and estradiol or VEGFA. Similarly, the expression of α-SMA on the surface of HUVECs treated with aldosterone, aldosterone with progesterone and estradiol were increased, while eplerenone supressed its expression. CONCLUSION Eplerenone inhibits renal angiogenesis by blocking the SGK-1/TGFβ signal transduction pathway, thereby inhibiting the phenotypic transformation of endothelial cells, slowing down renal fibrosis, and reducing kidney damage caused by pregnancy.
Collapse
Affiliation(s)
- Wenping Zhou
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Chang Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jieqi Niu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yunzhao Xiong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhen He
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hepeng Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Mengjuan Zhang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hongshuang Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qingyou Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiangting Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| | - Zheng Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| |
Collapse
|
3
|
Mu YF, Gao ZX, Mao ZH, Pan SK, Liu DW, Liu ZS, Wu P. Perspectives on the involvement of the gut microbiota in salt-sensitive hypertension. Hypertens Res 2024; 47:2351-2362. [PMID: 38877311 DOI: 10.1038/s41440-024-01747-y] [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: 11/13/2023] [Revised: 03/13/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024]
Abstract
Salt-sensitivity hypertension (SSH) is an independent predictor of cardiovascular event-related death. Despite the extensiveness of research on hypertension, which covers areas such as the sympathetic nervous system, the renin-angiotensin system, the vascular system, and the immune system, its pathogenesis remains elusive, with sub-optimal blood pressure control in patients. The gut microbiota is an important component of nutritional support and constitutes a barrier in the host. Long-term high salt intake can lead to gut microbiota dysbiosis and cause significant changes in the expression of gut microbiota-related metabolites. Of these metabolites, short chain fatty acids (SCFAs), trimethylamine oxide, amino acids, bile acids, and lipopolysaccharide are essential mediators of microbe-host crosstalk. These metabolites may contribute to the incidence and development of SSH via inflammatory, immune, vascular, and nervous pathways, among others. In addition, recent studies, including those on the histone deacetylase inhibitory mechanism of SCFAs and the blood pressure-decreasing effects of H2S via vascular activation, suggest that several proteins and factors in the classical pathway elicit their effects through multiple non-classical pathways. This review summarizes changes in the gut microbiota and its related metabolites in high-salt environments, as well as corresponding treatment methods for SSH, such as diet management, probiotic and prebiotic use, antibiotic use, and fecal transplantation, to provide new insights and perspectives for understanding SSH pathogenesis and the development of strategies for its treatment.
Collapse
Affiliation(s)
- Ya-Fan Mu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhong-Xiuzi Gao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zi-Hui Mao
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shao-Kang Pan
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dong-Wei Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China
- Henan Province Research Center for Kidney Disease, Zhengzhou, China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhang-Suo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| | - Peng Wu
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Institute of Nephrology, Zhengzhou University, Zhengzhou, China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| |
Collapse
|
4
|
Kim JH, Thiruvengadam R. Hypertension in an ageing population: Diagnosis, mechanisms, collateral health risks, treatments, and clinical challenges. Ageing Res Rev 2024; 98:102344. [PMID: 38768716 DOI: 10.1016/j.arr.2024.102344] [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: 07/20/2023] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Ageing population is considerably increasing worldwide, which is considered to reflect an improved quality of life. However, longevity in the human lifespan has increased the burden of late-life illnesses including cancer, neurodegeneration, and cardiovascular dysfunction. Of these, hypertension is the most common condition with huge health risks, with an increased prevalence among the elderly. In this review, we outline the current guidelines for defining hypertension and examine the detailed mechanisms underlying the relationship between hypertension and ageing-related outcomes, including sodium sensitivity, arterial stiffness, endothelial dysfunction, isolated systolic hypertension, white coat effect, and orthostatic hypertension. As hypertension-related collateral health risk increases among the elderly, the available management strategies are necessary to overcome the clinical treatment challenges faced among elderly population. To improve longevity and reduce adverse health effects, potential approaches producing crucial information into new era of medicine should be considered in the prevention and treatment of hypertension among elderly population. This review provides an overview of mechanisms underlying hypertension and its related collateral health risk in elderly population, along with multiple approaches and management strategies to improve the clinical challenges among elderly population.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, the Republic of Korea.
| | - Rekha Thiruvengadam
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, the Republic of Korea
| |
Collapse
|
5
|
Karabaeva RZ, Vochshenkova TA, Mussin NM, Albayev RK, Kaliyev AA, Tamadon A. Epigenetics of hypertension as a risk factor for the development of coronary artery disease in type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2024; 15:1365738. [PMID: 38836231 PMCID: PMC11148232 DOI: 10.3389/fendo.2024.1365738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Hypertension, a multifaceted cardiovascular disorder influenced by genetic, epigenetic, and environmental factors, poses a significant risk for the development of coronary artery disease (CAD) in individuals with type 2 diabetes mellitus (T2DM). Epigenetic alterations, particularly in histone modifications, DNA methylation, and microRNAs, play a pivotal role in unraveling the complex molecular underpinnings of blood pressure regulation. This review emphasizes the crucial interplay between epigenetic attributes and hypertension, shedding light on the prominence of DNA methylation, both globally and at the gene-specific level, in essential hypertension. Additionally, histone modifications, including acetylation and methylation, emerge as essential epigenetic markers linked to hypertension. Furthermore, microRNAs exert regulatory influence on blood pressure homeostasis, targeting key genes within the aldosterone and renin-angiotensin pathways. Understanding the intricate crosstalk between genetics and epigenetics in hypertension is particularly pertinent in the context of its interaction with T2DM, where hypertension serves as a notable risk factor for the development of CAD. These findings not only contribute to the comprehensive elucidation of essential hypertension but also offer promising avenues for innovative strategies in the prevention and treatment of cardiovascular complications, especially in the context of T2DM.
Collapse
Affiliation(s)
- Raushan Zh Karabaeva
- Gerontology Center, Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Tamara A. Vochshenkova
- Gerontology Center, Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Nadiar M. Mussin
- General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Rustam K. Albayev
- Gerontology Center, Medical Center Hospital of the President’s Affairs Administration of the Republic of Kazakhstan, Astana, Kazakhstan
| | - Asset A. Kaliyev
- General Surgery, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Amin Tamadon
- Department for Natural Sciences, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
- Department of Research and Development, PerciaVista R&D Co., Shiraz, Iran
| |
Collapse
|
6
|
Aljaibeji H, Heydarpour M, Stanton AM, Williams JS, Pojoga LH, Romero JR, Williams GH. Role of Raptor Gene Variants in Hypertension: Influence on Blood Pressure Independent of Salt Intake in White Population. Hypertension 2024; 81:1167-1177. [PMID: 38497230 PMCID: PMC11023780 DOI: 10.1161/hypertensionaha.123.22273] [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: 10/20/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND The mTOR (mechanistic target of rapamycin) is an essential regulator of fundamental biological processes. mTOR forms 2 distinct complexes, mTORC1 (mTOR complex 1) when it binds with RAPTOR (Regulatory-associated Protein of mTOR) and mTORC2 (mTOR complex 2) when it associates with RICTOR (Rapamycin-insesitive companion of mTOR). Due to the previous link between the mTOR pathway, aldosterone, and blood pressure (BP), we anticipated that variants in the mTOR complex might be associated with salt-sensitive BP. METHODS BP and other parameters were assessed after a one-week liberal Na+ (200 mmol/d) and a one-week restricted Na+ (10 mmol/d) diet in 608 White subjects from the Hypertensive Pathotype cohort, single-nucleotide variants in MTOR, RPTOR, and RICTOR genes were obtained for candidate genes analyses. RESULTS The analysis revealed a significant association between a single nucleotide variants within the RPTOR gene and BP. Individuals carrying the RPTOR rs9901846 homozygous risk allele (AA) and heterozygous risk allele (GA) exhibited a 5 mm Hg increase in systolic BP on a liberal diet compared with nonrisk allele individuals (GG), but only in women. This single nucleotide variants effect was more pronounced on the restricted diet and present in both sexes, with AA carriers having a 9 mm Hg increase and GA carriers having a 5 mm Hg increase in systolic BP compared with GG. Interestingly, there were no significant associations between MTOR or RICTOR gene variants and BP. CONCLUSIONS The RPTOR gene variation is associated with elevated BP in White participants, regardless of salt intake, specifically in females.
Collapse
Affiliation(s)
- Hayat Aljaibeji
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Mahyar Heydarpour
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Ana Maria Stanton
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Jonathan S Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Luminita H Pojoga
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Jose R Romero
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| | - Gordon H Williams
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA (H.A., M.H., A.M.S., J.S.W., L.H.P., J.R.R., G.H.W.)
| |
Collapse
|
7
|
Shilole JN, Omari RB, Ruhighira JJ, Khamis AG, Ntwenya JE. Adherence to Lifestyle Recommendations among Adults Attending Hypertension Clinics in Selected Hospitals in Tanzania: A Cross-Sectional Study. East Afr Health Res J 2024; 8:25-31. [PMID: 39234338 PMCID: PMC11371004 DOI: 10.24248/eahrj.v8i1.745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 03/28/2024] [Indexed: 09/06/2024] Open
Abstract
Background and Aims Hypertension is the first contributor to the deaths caused by non-communicable diseases (NCDs) worldwide. A change of lifestyle is recommended as an equal-first-line approach for controlling hypertension. However, the burden of uncontrolled hypertension remains high. This article describes the level of adherence to recommended lifestyle modifications among hypertensive patients in Tanzania. Methods The research was carried out from June to September 2020 using a cross-sectional study that involved an interviewer-administered questionnaire with 311 participants. These were patients with hypertension (> 18 years old) who were randomly selected from patients attending clinics during the study period. The lifestyle behaviours were assessed using the WHO Steps survey standard questionnaire. SPSS, version 26, was used to enter and analyse the data. Results The mean age of hypertensive patients was 53.6 ± 7.5 years. Females were 58.8%. Only 17.7% had good compliance with the recommended lifestyle behaviours related to hypertension. Regular physical activities had 37.9% adherence, 99% adhered to non-smoking, 94.2% adhered to moderation of alcohol consumption, and 22.2% adhered to the consumption of fruits and vegetables. Patients with adequate knowledge were two times more likely to comply with the WHO recommended lifestyle behaviours (aOR=2.32; 95% confidence interval [CI], 1.082 to 3.471; P= .05]. Conclusion Most patients with hypertension had poor lifestyle behaviours for the management of hypertension, with varying level of adherence to the recommended life style changes.
Collapse
Affiliation(s)
| | | | | | - Ahmed Gharib Khamis
- Department of Epidemiology and Biostatistics; Muhimbili University of Health and Allied Sciences, Tanzania
| | | |
Collapse
|
8
|
Xu C, Zhu J, Gong G, Guo L, Zhang Y, Zhang Z, Ma C. Anthocyanin attenuates high salt-induced hypertension via inhibiting the hyperactivity of the sympathetic nervous system. Clin Exp Hypertens 2023; 45:2233717. [PMID: 37454306 DOI: 10.1080/10641963.2023.2233717] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/22/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Anthocyanin plays a protective role in cardiovascular disease through antioxidant effect. Whether anthocyanin can reduce salt-induced hypertension and the related mechanisms remain unclear. METHODS Chronic infusion of vehicle (artificial cerebrospinal fluid, aCSF, 0.4 μL/h) or anthocyanin (10 mg/kg, 0.4 μL/h) into bilateral paraventricular nucleus (PVN) of Sprague-Dawley rats was performed. Then, the rats were fed a high salt diet (8% NaCl, HS) or normal salt diet (0.9%, NaCl, NS) for 4 weeks. RESULTS High salt diet induced an increase in blood pressure and peripheral sympathetic nerve activity (increased LF/HF and decreased SDNN and RMSSD), which was accompanied by increased reactive oxygen species (ROS) production and angiotensin II type-1 receptor (AT1R) expression and function in the PVN. Moreover, the NOD-like receptor protein 3 (NLRP3) and related inflammatory proteins (caspase-1) expression, the pro-inflammatory cytokine levels including IL-1β and TNF-α were higher in PVN of rats with a high salt diet. Bilateral PVN infusion of anthocyanin attenuated NLRP3-dependent inflammation (NLRP3, caspase-1, IL-1β and TNF-α) and ROS production, reduced AT1R expression and function in PVN and lowered peripheral sympathetic nerve activity and blood pressure in rats with salt-induced hypertension. CONCLUSIONS Excessive salt intake activates NLRP3-dependent inflammation and oxidative stress and increased AT1R expression and function in the PVN. Bilateral PVN infusion of anthocyanin lowers peripheral sympathetic nerve activity and blood pressure in rats with salt-induced hypertension by improvement of expression and function of AT1R in the PVN through inhibiting NLRP3 related inflammatory and oxidative stress.
Collapse
Affiliation(s)
- Chunmei Xu
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jun Zhu
- Department of Cardiology, Shanghai Hospital, Chongqing, China
| | - Guangyuan Gong
- Department of Intensive Care Unit, Qijiang People's Hospital, Chongqing, China
| | - Li Guo
- Department of Endocrinology, The Southwest Hospital of Army Medical University, Chongqing, China
| | - Ye Zhang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Ziyue Zhang
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
| | - Chunlan Ma
- Department of Cardiology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
9
|
Silverio A, Bossone E, Parodi G, Scudiero F, Di Maio M, Vriz O, Bellino M, Zito C, Provenza G, Iuliano G, Cristiano M, Novo G, Mauro C, Rigo F, Innelli P, Salerno-Uriarte J, Cameli M, Tremiterra G, Vecchione C, Antonini-Canterin F, Galasso G, Citro R. Arterial hypertension in patients with takotsubo syndrome: prevalence, long-term outcome, and secondary preventive strategies: a report from the Takotsubo Italian Network register. Eur J Prev Cardiol 2023; 30:1998-2005. [PMID: 37463434 DOI: 10.1093/eurjpc/zwad237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/18/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023]
Abstract
AIMS The aim of this study was to investigate the long-term outcome of takotsubo syndrome (TTS) patients with and without hypertension (HT) and to evaluate the effectiveness of treatment with beta-blockers (BBs) and/or renin-angiotensin-aldosterone system inhibitors (RAASi). METHODS AND RESULTS The study population includes a register-based, multicentre cohort of consecutive patients with TTS, divided into two groups according to the history of HT. Further stratification was performed for BB/RAASi prescription at discharge. The primary outcome was the composite of all-cause death and TTS recurrence at the longest available follow-up. The propensity score weighting technique was used to account for potential confounding. In the overall population (903 patients, mean age 70 ± 11 years), HT was reported in 66% of cases. At a median 2-year follow-up, there was no difference in the risk of the primary composite outcome between patients with and without HT. The adjusted Cox regression analysis showed a significantly lower risk for the primary outcome [adjusted hazard ratio (aHR): 0.69; 95% confidence interval (CI): 0.49-0.99] in patients who received BB vs. those who did not. Renin-angiotensin-aldosterone system inhibitors treatment was not associated with the primary study outcome. The lower risk for the primary outcome with BB treatment was confirmed in patients with HT (aHR: 0.37; 95% CI: 0.24-0.56) but not in patients without (aHR: 1.83; 95% CI: 0.92-3.64; Pinteraction < 0.001). CONCLUSION In this TTS study, HT did not affect the long-term risk of adverse events but increased the probability of benefit from BB treatment after discharge. Owing to the favourable outcome impact of BB prescription in TTS patients with HT, a tailored pharmacological therapy should be considered in this cohort.
Collapse
Affiliation(s)
- Angelo Silverio
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Eduardo Bossone
- Division of Cardiology, Antonio Cardarelli Hospital, Naples, Italy
| | - Guido Parodi
- Department of Cardiology, ASL4 Liguria, Lavagna, Italy
| | | | - Marco Di Maio
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Olga Vriz
- Heart Centre, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- College of Medicine, Al Faisal University, Riyadh, Saudi Arabia
| | - Michele Bellino
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Concetta Zito
- Department of Clinical and Experimental Medicine, Section of Cardiology, University of Messina, University Hospital 'Policlinico G. Martino', Messina, Italy
| | - Gennaro Provenza
- Division of Cardiology, Cardiovascular and Thoracic Department, San Giovanni di Dio e Ruggi d'Aragona University Hospital, Heart Tower Room 807, Largo Città d'Ippocrate, Salerno 84131, Italy
| | - Giuseppe Iuliano
- Division of Cardiology, Cardiovascular and Thoracic Department, San Giovanni di Dio e Ruggi d'Aragona University Hospital, Heart Tower Room 807, Largo Città d'Ippocrate, Salerno 84131, Italy
| | - Mario Cristiano
- Division of Cardiology, Cardiovascular and Thoracic Department, San Giovanni di Dio e Ruggi d'Aragona University Hospital, Heart Tower Room 807, Largo Città d'Ippocrate, Salerno 84131, Italy
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Ciro Mauro
- Division of Cardiology, Antonio Cardarelli Hospital, Naples, Italy
| | - Fausto Rigo
- Cardiology Department, Ospedale dell'Angelo Mestre-Venice, Venice, Italy
| | - Pasquale Innelli
- Department of Cardiovascular Imaging, San Carlo Hospital, Potenza, Italy
| | | | - Matteo Cameli
- Division of Cardiology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Giuliana Tremiterra
- Healtcare Management, San Giovanni di Dio e Ruggi d'Aragona University Hospital, Salerno, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
- Vascular Physiopathology Unit, IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Isernia, Italy
| | - Francesco Antonini-Canterin
- Cardiology Unit, High Specialization Rehabilitation Hospital Motta di Livenza, Motta di Livenza, Treviso, Italy
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry, University of Salerno, Baronissi, Salerno, Italy
| | - Rodolfo Citro
- Division of Cardiology, Cardiovascular and Thoracic Department, San Giovanni di Dio e Ruggi d'Aragona University Hospital, Heart Tower Room 807, Largo Città d'Ippocrate, Salerno 84131, Italy
- Vascular Physiopathology Unit, IRCCS Neuromed, Via Atinense 18, 86077 Pozzilli, Isernia, Italy
| |
Collapse
|
10
|
Yang P, Rooney MR, Wallace AS, Kim H, Echouffo-Tcheugui JB, McEvoy JW, Ndumele C, Christenson RH, Selvin E, Rebholz CM. Associations between diet quality and NT-proBNP in U.S. adults, NHANES 1999-2004. Am J Prev Cardiol 2023; 16:100528. [PMID: 37601625 PMCID: PMC10432600 DOI: 10.1016/j.ajpc.2023.100528] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/22/2023] Open
Abstract
Objective N-terminal pro-brain-type natriuretic peptide (NT-proBNP) is a marker of cardiac wall stress and is a predictor of cardiovascular disease. Higher diet quality is associated with lower risk of cardiovascular disease. The association between diet quality and subclinical cardiovascular disease assessed by NT-proBNP is uncharacterized. We investigated the associations between diet quality, using Healthy Eating Index-2015 (HEI-2015), and NT-proBNP from the National Health and Nutrition Examination Survey (NHANES) 1999-2004. Methods We included 9,782 adults from NHANES 1999-2004 without self-reported cardiovascular disease. The HEI-2015 ranges from 0 to 100, with higher scores indicating better diet quality. The HEI-2015 was categorized into sex-specific quintiles. Regression models were used to quantify associations between the overall HEI-2015 score and its 13 components with log-transformed NT-proBNP. The beta coefficients were converted to percent differences. Results Among 9,782 participants, mean age was 45 years, 48% were men, and 72% were non-Hispanic White adults. After adjusting for sociodemographic characteristics, lifestyle factors, and medical history, those in the highest vs. lowest HEI-2015 quintile had an 8.5% (95% CI: -14.6% to -2.0%) lower NT-proBNP level. There was a dose-response association between HEI-2015 and NT-proBNP (P value for trend = 0.01). Each 1-unit higher in sodium and added sugars score indicating lower intake was associated with lower NT-proBNP by 7.7% (95% CI: -12.8% to -2.2%) and 6.5% (95% CI: -12.0% to -0.7%), respectively. Conclusion Higher diet quality, especially lower intakes of sodium and added sugars, was associated with lower serum levels of NT-proBNP.
Collapse
Affiliation(s)
- Ping Yang
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Mary R. Rooney
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Amelia S. Wallace
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Hyunju Kim
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Justin B. Echouffo-Tcheugui
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John W. McEvoy
- National Institute for Prevention and Cardiovascular Health, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Chiadi Ndumele
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert H Christenson
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Elizabeth Selvin
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Casey M. Rebholz
- Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
11
|
Petersen JCG, Jonassen TEN, Holstein-Rathlou NH, Petersen LG, Sorensen CM. Dynamic changes in renal sodium handling during sympathetic stimulation in healthy human males. Auton Neurosci 2023; 250:103131. [PMID: 37984257 DOI: 10.1016/j.autneu.2023.103131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/27/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
The temporal response of changes in renal sodium reabsorption during increased renal sympathetic nerve activity has not been investigated. Central hypovolemia by application of lower-body negative-pressure (LBNP) elicits baroreceptor mediated sympathetic reflexes to maintain arterial blood pressure. We hypothesized, that during 90 min LBNP, the renal sodium retention would increase rapidly, remain increased during intervention, and return to baseline immediately after end of intervention. METHODS 30 young, healthy, sodium loaded, non-obese males were exposed to -15 mmHg LBNP, -30 mmHg LBNP, -15 mmHg LBNP + renin blockade or time-control (0 mmHg LBNP) for 90 min. Urine was collected every 15 min during 90 min of intervention and 60 min of recovery to identify a possible relation between time of intervention and renal response. RESULTS All intervention groups exhibited a comparable reduction in distal sodium excretion at the end of the intervention (P = 0.46 between groups; -15 mmHg: -3.1 ± 0.9 %, -30 mmHg: -2.9 ± 0.6 %, -15 mmHg + aslikiren: -1.8 ± 0.6 %). -15 mmHg+Aliskiren resulted in a slower onset, but all groups exhibited a continued reduction in sodium excretion after 1 h of recovery despite return to baseline of renin, aldosterone, diuresis and cardiovascular parameters. CONCLUSION Sympathetic stimulation for 90 min via LBNP at -30 mmHg LBNP compared to -15 mmHg did not result in a greater response in fractional Na+ excretion, suggesting that additional baroreceptor unloading did not cause further increases in renal sodium reabsorption. Changes in distal Na+ excretion were linear with respect to time (dose) of intervention, but seem to exhibit a saturation-like effect at a level around 4 %. The lack of recovery after 1 h is also a new finding that warrants further investigation.
Collapse
Affiliation(s)
- J C G Petersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - T E N Jonassen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - N-H Holstein-Rathlou
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - L G Petersen
- Department of Aeronautics and Astronautics, Massachusetts Institute of Technology, Cambridge, USA
| | - C M Sorensen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
12
|
Buncha V, Cherezova A, Alexander S, Baranovskaya I, Coleman KA, Cherian-Shaw M, Brands MW, Sullivan JC, O'Connor PM, Mamenko M. Aldosterone Antagonism Is More Effective at Reducing Blood Pressure and Excessive Renal ENaC Activity in AngII-Infused Female Rats Than in Males. Hypertension 2023; 80:2196-2208. [PMID: 37593894 PMCID: PMC10528186 DOI: 10.1161/hypertensionaha.123.21287] [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: 03/27/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND AngII (angiotensin II)-dependent hypertension causes comparable elevations of blood pressure (BP), aldosterone levels, and renal ENaC (epithelial Na+ channel) activity in male and female rodents. Mineralocorticoid receptor (MR) antagonism has a limited antihypertensive effect associated with insufficient suppression of renal ENaC in male rodents with AngII-hypertension. While MR blockade effectively reduces BP in female mice with salt-sensitive and leptin-induced hypertension, MR antagonism has not been studied in female rodents with AngII-hypertension. We hypothesize that overstimulation of renal MR signaling drives redundant ENaC-mediated Na+ reabsorption and BP increase in female rats with AngII-hypertension. METHODS We employ a combination of physiological, pharmacological, biochemical, and biophysical approaches to compare the effect of MR inhibitors on BP and ENaC activity in AngII-infused male and female Sprague Dawley rats. RESULTS MR blockade markedly attenuates AngII-hypertension in female rats but has only a marginal effect in males. Spironolactone increases urinary sodium excretion and urinary sodium-to-potassium ratio in AngII-infused female, but not male, rats. The expression of renal MR and HSD11β2 (11β-hydroxysteroid dehydrogenase type 2) that determines the availability of MR to aldosterone is significantly higher in AngII-infused female rats than in males. ENaC activity is ≈2× lower in spironolactone-treated AngII-infused female rats than in males. Reduced ENaC activity in AngII-infused female rats on spironolactone correlates with increased interaction with ubiquitin ligase Nedd4-2 (neural precursor cell expressed developmentally down-regulated protein 4-2), targeting ENaC for degradation. CONCLUSIONS MR-ENaC axis is the primary determinant of excessive renal sodium reabsorption and an attractive antihypertensive target in female rats with AngII-hypertension, but not in males.
Collapse
Affiliation(s)
- Vadym Buncha
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Alena Cherezova
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Sati Alexander
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Irina Baranovskaya
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Kathleen A Coleman
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia, Augusta University
| | | | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia, Augusta University
| | - Mykola Mamenko
- Department of Physiology, Medical College of Georgia, Augusta University
| |
Collapse
|
13
|
Masenga SK, Kirabo A. Hypertensive heart disease: risk factors, complications and mechanisms. Front Cardiovasc Med 2023; 10:1205475. [PMID: 37342440 PMCID: PMC10277698 DOI: 10.3389/fcvm.2023.1205475] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
Hypertensive heart disease constitutes functional and structural dysfunction and pathogenesis occurring primarily in the left ventricle, the left atrium and the coronary arteries due to chronic uncontrolled hypertension. Hypertensive heart disease is underreported and the mechanisms underlying its correlates and complications are not well elaborated. In this review, we summarize the current understanding of hypertensive heart disease, we discuss in detail the mechanisms associated with development and complications of hypertensive heart disease especially left ventricular hypertrophy, atrial fibrillation, heart failure and coronary artery disease. We also briefly highlight the role of dietary salt, immunity and genetic predisposition in hypertensive heart disease pathogenesis.
Collapse
Affiliation(s)
- Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Cam-Pus, Livingstone, Zambia
- School of Medicine, University of Zambia, Lusaka, Zambia
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, TN, United States
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Centre, Nashville, TN, United States
| |
Collapse
|
14
|
Purinoceptor: a novel target for hypertension. Purinergic Signal 2023; 19:185-197. [PMID: 35181831 PMCID: PMC9984596 DOI: 10.1007/s11302-022-09852-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/08/2022] [Indexed: 12/11/2022] Open
Abstract
Hypertension is the leading cause of morbidity and mortality globally among all cardiovascular diseases. Purinergic signalling plays a crucial role in hypertension through the sympathetic nerve system, neurons in the brain stem, carotid body, endothelium, immune system, renin-angiotensin system, sodium excretion, epithelial sodium channel activity (ENaC), and renal autoregulation. Under hypertension, adenosine triphosphate (ATP) is released as a cotransmitter from the sympathetic nerve. It mediates vascular tone mainly through P2X1R activation on smooth muscle cells and activation of P2X4R and P2YR on endothelial cells and also via interaction with other purinoceptors, showing dual effects. P2Y1R is linked to neurogenic hypertension. P2X7R and P2Y11R are potential targets for immune-related hypertension. P2X3R located on the carotid body is the most promising novel therapeutic target for hypertension. A1R, A2AR, A2BR, and P2X7R are all related to renal autoregulation, which contribute to both renal damage and hypertension. The main focus is on the evidence addressing the involvement of purinoceptors in hypertension and therapeutic interventions.
Collapse
|
15
|
Dietary salt intake predicts future development of metabolic syndrome in the general population. Hypertens Res 2023; 46:236-243. [PMID: 36229525 DOI: 10.1038/s41440-022-01035-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 02/03/2023]
Abstract
Excessive dietary salt consumption is one of the most important risk factors for hypertension. Metabolic disorders often coexist with hypertension, and excess salt intake has been reported to underlie metabolic disorders, such as insulin resistance. Therefore, we tested the hypothesis that excessive dietary salt causes metabolic syndrome in the general population. In total, 13886 subjects who participated in our medical checkup were enrolled, and salt intake was assessed using a spot urine sample. The characteristics of participants with metabolic syndrome (n = 1630) were examined at baseline, and then participants without metabolic syndrome (n = 12256) were followed up with the endpoint being the development of metabolic syndrome. The average estimated salt intake in our participants was 8.72 ± 1.93 g/day. A significant association between salt intake and metabolic syndrome was obtained from the logistic regression analysis, and salt intake increased as the number of metabolic disorders in an individual increased at baseline (P < 0.001). During the median follow-up period of 52 months, 1669 participants developed metabolic syndrome. Kaplan-Meier analysis demonstrated an increased risk of metabolic syndrome across quartiles of baseline salt intake (log-rank, P < 0.001). In the Cox proportional hazard regression analysis where salt intake was taken as a continuous variable, salt intake at baseline was an independent predictor of developing metabolic syndrome. These results suggest that excessive salt intake is significantly associated with the development of metabolic syndrome in the general population. Salt may play an important role in the development of metabolic disorders and hypertension.
Collapse
|
16
|
Molecular Mechanisms of Na-Cl Cotransporter in Relation to Hypertension in Chronic Kidney Disease. Int J Mol Sci 2022; 24:ijms24010286. [PMID: 36613730 PMCID: PMC9820686 DOI: 10.3390/ijms24010286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic kidney disease (CKD) is a common clinical disease with an increasing incidence, affecting 10 to 15% of the world's population. Hypertension is the most common and modifiable risk factor for preventing adverse cardiovascular outcomes in patients with CKD. A survey from developed countries shows that 47% of hypertensive patients over the age of 20 have uncontrolled blood pressure (BP), and the control rate is even lower in developing countries. CKD is both a common cause of uncontrolled hypertension and a risk factor for altered sequelae. In particular, studies have demonstrated that abnormal blood-pressure patterns in CKD patients, such as non-dipping-blood-pressure patterns, are associated with a significantly increased risk of cardiovascular (CV) disease. The distal convoluted tubule (DCT) is a region of the kidney, and although only 5-10% of the sodium (Na+) filtered by the glomerulus is reabsorbed by DCT, most studies agree that Na-Cl cotransporter (NCC) in human, rabbit, mouse, and rat kidneys is the most important route of sodium reabsorption across the DCT for maintaining the homeostasis of sodium. The regulation of NCC involves a large and complex network structure, including certain physiological factors, kinases, scaffold proteins, transporter phosphorylation, and other aspects. This regulation network includes various levels. Naturally, cross-talk between the components of this system must occur in order to relay the important signals to the transporter to play its role. Knowledge of the mechanisms regulating NCC activation is critical for understanding and treating hypertension and CKD. Previous studies from our laboratory have investigated the mechanisms through which NCC is activated in several different models. In the following sections, we review the literature on the mechanisms of NCC in relation to hypertension in CKD.
Collapse
|
17
|
Xie Y, Qi H, Peng W, Li B, Wen F, Zhang F, Zhang L. Higher Potassium Intake and Lower Sodium Intake May Help in Reducing CVD Risk by Lowering Salt Sensitivity of Blood Pressure in the Han Chinese Population. Nutrients 2022; 14:nu14204436. [PMID: 36297119 PMCID: PMC9607620 DOI: 10.3390/nu14204436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 03/10/2023] Open
Abstract
Sodium (Na) reduction with a parallel supplemental potassium (K) intake can prevent cardiovascular diseases (CVDs). The relationship of the urinary Na/K ratio and salt sensitivity of blood pressure (SSBP) with CVDs is not clearly explained. We assumed that the SSBP mediates the relationship between the Na/K ratio and CVDs. In total, 2055 subjects who had 24 h urine collected and SSBP determined were included in this study. CVD risk was estimated using the China-PAR equation. MediationMultivariate logistic regression was used to explore the associations between the Na/K ratio or SSBP with CVD risk. Mediation analysis using a logistic regression model was performed. Both the urinary Na/K ratio and SSBP were related to the estimated CVD risk (p < 0.05). The mediation analysis found that SSBP mediated approximately 12% of the association between Na/K ratio and CVD risk. Our findings indicate that higher K intake and lower Na intake may help in preventing CVD risk by reducing SSBP risk in individuals with normotension or stage-one hypertension.
Collapse
Affiliation(s)
- Yunyi Xie
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Han Qi
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Wenjuan Peng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Bingxiao Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Fuyuan Wen
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Fengxu Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Ling Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
- Correspondence: ; Tel.: +86-10-83911777
| |
Collapse
|
18
|
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: 8] [Impact Index Per Article: 2.7] [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
|
19
|
Hogas M, Statescu C, Padurariu M, Ciobica A, Bilha SC, Haisan A, Timofte D, Hogas S. Salt, Not Always a Cardiovascular Enemy? A Mini-Review and Modern Perspective. Medicina (B Aires) 2022; 58:medicina58091175. [PMID: 36143852 PMCID: PMC9504547 DOI: 10.3390/medicina58091175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Dietary salt intake is a long-debated issue. Increased sodium intake is associated with high blood pressure, leading to salt-sensitive hypertension. Excessive salt intake leads to arterial stiffness in susceptible individuals via impaired nitric oxide action and increased endothelin-1 expression, overactivity of the renal sympathetic nervous system and also via aldosterone-independent activation of the mineralocorticoid receptor. Salt restriction in such individuals reduces blood pressure (BP) values. The optimal level of salt restriction that leads to improved cardiovascular outcomes is still under debate. Current BP and dietary guidelines recommend low sodium intake for the general population. However, a specific category of patients does not develop arterial hypertension in response to sodium loading. In addition, recent research demonstrates the deleterious effects of aggressive sodium restriction, even in heart failure patients. This mini review discusses current literature data regarding the advantages and disadvantages of salt restriction and how it impacts the overall health status.
Collapse
Affiliation(s)
- Mihai Hogas
- Physiology Department, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii 16, 700115 Iasi, Romania
| | - Cristian Statescu
- Cardiology Department, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii 16, 700115 Iasi, Romania
| | - Manuela Padurariu
- Psychiatry Department, “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii 16, 700115 Iasi, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, No 11, 700115 Iasi, Romania
- Academy of Romanian Scientists, Splaiul Independentei Nr. 54, Sector 5, 050094 Bucuresti, Romania
- Center of Biomedical Research, Romanian Academy, B dul Carol I, No 8, 700115 Iasi, Romania
| | - Stefana Catalina Bilha
- Endocrinology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (S.C.B.); (A.H.)
| | - Anca Haisan
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Correspondence: (S.C.B.); (A.H.)
| | - Daniel Timofte
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Simona Hogas
- Nephrology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
20
|
Zhao Y, Li L, Lu Z, Hu Y, Zhang H, Sun F, Li Q, He C, Shu W, Wang L, Cao T, Luo Z, Yan Z, Liu D, Gao P, Zhu Z. Sodium-Glucose Cotransporter 2 Inhibitor Canagliflozin Antagonizes Salt-Sensitive Hypertension Through Modifying Transient Receptor Potential Channels 3 Mediated Vascular Calcium Handling. J Am Heart Assoc 2022; 11:e025328. [PMID: 35904193 PMCID: PMC9375510 DOI: 10.1161/jaha.121.025328] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Salt-sensitive hypertension is highly prevalent and associated with cardiorenal damage. Large clinical trials have demonstrated that SGLT2 (sodium-glucose cotransporter 2) inhibitors exert hypotensive effect and cardiorenal protective benefits in patients with hypertension with and without diabetes. However, the underlying mechanism remains elusive. Methods and Results Dahl salt-sensitive rats and salt-insensitive controls were fed with 8% high-salt diet and some of them were treated with canagliflozin. The blood pressure, urinary sodium excretion, and vascular function were detected. Transient receptor potential channel 3 (TRPC3) knockout mice were used to explain the mechanism. Canagliflozin treatment significantly reduced high-salt-induced hypertension and this effect was not totally dependent on urinary sodium excretion in salt-sensitive hypertensive rats. Assay of vascular function and proteomics showed that canagliflozin significantly inhibited vascular cytoplasmic calcium increase and vasoconstriction in response to high-salt diet. High salt intake increased vascular expression of TRPC3 in salt-sensitive rats, which could be alleviated by canagliflozin treatment. Overexpression of TRPC3 mimicked salt-induced vascular cytosolic calcium increase in vitro and knockout of TRPC3 erased the antihypertensive effect of canagliflozin. Mechanistically, high-salt-induced activation of NCX1 (sodium-calcium exchanger 1) reverse mode increased cytoplasmic calcium level and vasoconstriction, which required TRPC3, and this process could be blocked by canagliflozin. Conclusions We define a previously unrecognized role of TRPC3/NCX1 mediated vascular calcium dysfunction in the development of high-salt-induced hypertension, which can be improved by canagliflozin treatment. This pathway is potentially a novel therapeutic target to antagonize salt-sensitive hypertension.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Li Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zongshi Lu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Yingru Hu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Hexuan Zhang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Fang Sun
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Qiang Li
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Chengkang He
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Wentao Shu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Lijuan Wang
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Tingbing Cao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhidan Luo
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhencheng Yan
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Daoyan Liu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Peng Gao
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| | - Zhiming Zhu
- Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital Army Medical University, Chongqing Institute of Hypertension Chongqing China
| |
Collapse
|
21
|
Plasma B-type natriuretic peptide level as a possible predictor of dietary salt intake. Hypertens Res 2022; 45:1134-1135. [PMID: 35581499 DOI: 10.1038/s41440-022-00938-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 11/09/2022]
|
22
|
Xu JF, Xia J, Wan Y, Yang Y, Wu JJ, Peng C, Ao H. Vasorelaxant Activities and its Underlying Mechanisms of Magnolia Volatile Oil on Rat Thoracic Aorta Based on Network Pharmacology. Front Pharmacol 2022; 13:812716. [PMID: 35308213 PMCID: PMC8926352 DOI: 10.3389/fphar.2022.812716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Objective: Magnolia volatile oil (MVO) is a mixture mainly containing eudesmol and its isomers. This study was to investigate the vasorelaxant effects and the underlying mechanism of MVO in rat thoracic aortas. Method: The present study combined gas chromatography–mass spectrometry (GC-MS) and network pharmacology analysis with in vitro experiments to clarify the mechanisms of MVO against vessel contraction. A compound–target network, compound–target–disease network, protein–protein interaction network, compound–target–pathway network, gene ontology, and pathway enrichment for hypertension were applied to identify the potential active compounds, drug targets, and pathways. Additionally, the thoracic aortic rings with or without endothelium were prepared to explore the underlying mechanisms. The roles of the PI3K-Akt-NO pathways, neuroreceptors, K+ channels, and Ca2+ channels on the vasorelaxant effects of MVO were evaluated through the rat thoracic aortic rings. Results: A total of 29 compounds were found in MVO, which were identified by GC-MS, of which 21 compounds with a content of more than 0.1% were selected for further analysis. The network pharmacology research predicted that beta-caryophyllene, palmitic acid, and (+)-β-selinene might act as the effective ingredients of MVO for the treatment of hypertension. Several hot targets, mainly involving TNF, CHRM1, ACE, IL10, PTGS2, REN, and F2, and pivotal pathways, such as the neuroactive ligand–receptor interaction, the calcium signaling pathway, and the PI3K-Akt signaling, were responsible for the vasorelaxant effect of MVO. As expected, MVO exerted a vasorelaxant effect on the aortic rings pre-contracted by KCl and phenylephrine in an endothelium-dependent and non-endothelium-dependent manner. Importantly, a pre-incubation with indomethacin (Indo), N-nitro-L-arginine methyl ester, methylene blue, wortmannin, and atropine sulfate as well as 4-aminopyridione diminished MVO-induced vasorelaxation, suggesting that the activation of the PI3K-Akt-NO pathway and KV channel were involved in the vasorelaxant effect of MVO, which was consistent with the results of the Kyoto Encyclopedia of Genes and the Genomes. Additionally, MVO could significantly inhibit Ca2+ influx resulting in the contraction of aortic rings, revealing that the inhibition of the calcium signaling pathway exactly participated in the vasorelaxant activity of MVO as predicted by network pharmacology. Conclusion: MVO might be a potent treatment of diseases with vascular dysfunction like hypertension. The underlying mechanisms were related to the PI3K-Akt-NO pathway, KV pathway, as well as Ca2+ channel, which were predicted by the network pharmacology and verified by the experiments in vitro. This study based on network pharmacology provided experimental support for the clinical application of MVO in the treatment of hypertension and afforded a novel research method to explore the activity and mechanism of traditional Chinese medicine.
Collapse
Affiliation(s)
- Jin-Feng Xu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Xia
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Wan
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Yang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiao-Jiao Wu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Cheng Peng, ; Hui Ao,
| | - Hui Ao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Cheng Peng, ; Hui Ao,
| |
Collapse
|
23
|
Labban M, Itani MM, Maaliki D, Nasreddine L, Itani HA. The Sweet and Salty Dietary Face of Hypertension and Cardiovascular Disease in Lebanon. Front Physiol 2022; 12:802132. [PMID: 35153813 PMCID: PMC8835350 DOI: 10.3389/fphys.2021.802132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
According to the World Health Organization (WHO), an estimated 1.28 billion adults aged 30–79 years worldwide have hypertension; and every year, hypertension takes 7.6 million lives. High intakes of salt and sugar (mainly fructose from added sugars) have been linked to the etiology of hypertension, and this may be particularly true for countries undergoing the nutrition transition, such as Lebanon. Salt-induced hypertension and fructose-induced hypertension are manifested in different mechanisms, including Inflammation, aldosterone-mineralocorticoid receptor pathway, aldosterone independent mineralocorticoid receptor pathway, renin-angiotensin system (RAS), sympathetic nervous system (SNS) activity, and genetic mechanisms. This review describes the evolution of hypertension and cardiovascular diseases (CVDs) in Lebanon and aims to elucidate potential mechanisms where salt and fructose work together to induce hypertension. These mechanisms increase salt absorption, decrease salt excretion, induce endogenous fructose production, activate fructose-insulin-salt interaction, and trigger oxidative stress, thus leading to hypertension. The review also provides an up-to-date appraisal of current intake levels of salt and fructose in Lebanon and their main food contributors. It identifies ongoing salt and sugar intake reduction strategies in Lebanon while acknowledging the country’s limited scope of regulation and legislation. Finally, the review concludes with proposed public health strategies and suggestions for future research, which can reduce the intake levels of salt and fructose levels and contribute to curbing the CVD epidemic in the country.
Collapse
Affiliation(s)
| | - Maha M Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Dina Maaliki
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Lara Nasreddine
- Vascular Medicine Program, American University of Beirut Medical Center, Beirut, Lebanon.,Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Hana A Itani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Vascular Medicine Program, American University of Beirut Medical Center, Beirut, Lebanon.,Adjunct Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
24
|
Kidney Damage Caused by Obesity and Its Feasible Treatment Drugs. Int J Mol Sci 2022; 23:ijms23020747. [PMID: 35054932 PMCID: PMC8775419 DOI: 10.3390/ijms23020747] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 02/07/2023] Open
Abstract
The rapid growth of obesity worldwide has made it a major health problem, while the dramatic increase in the prevalence of obesity has had a significant impact on the magnitude of chronic kidney disease (CKD), especially in developing countries. A vast amount of researchers have reported a strong relationship between obesity and chronic kidney disease, and obesity can serve as an independent risk factor for kidney disease. The histological changes of kidneys in obesity-induced renal injury include glomerular or tubular hypertrophy, focal segmental glomerulosclerosis or bulbous sclerosis. Furthermore, inflammation, renal hemodynamic changes, insulin resistance and lipid metabolism disorders are all involved in the development and progression of obesity-induced nephropathy. However, there is no targeted treatment for obesity-related kidney disease. In this review, RAS inhibitors, SGLT2 inhibitors and melatonin would be presented to treat obesity-induced kidney injury. Furthermore, we concluded that melatonin can protect the kidney damage caused by obesity by inhibiting inflammation and oxidative stress, revealing its therapeutic potential.
Collapse
|
25
|
Ding R, Xiao Z, Jiang Y, Yang Y, Ji Y, Bao X, Xing K, Zhou X, Zhu S. Calcitriol ameliorates damage in high-salt diet-induced hypertension: Evidence of communication with the gut-kidney axis. Exp Biol Med (Maywood) 2021; 247:624-640. [PMID: 34894804 DOI: 10.1177/15353702211062507] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Several studies have established a link between high-salt diet, inflammation, and hypertension. Vitamin D supplementation has shown anti-inflammatory effects in many diseases; gut microbiota is also associated with a wide variety of cardiovascular diseases, but potential role of vitamin D and gut microbiota in high-salt diet-induced hypertension remains unclear. Therefore, we used rats with hypertension induced by a high-salt diet as the research object and analyzed the transcriptome of their tissues (kidney and colon) and gut microbiome to conduct an overall analysis of the gut-kidney axis. We aimed to confirm the effects of high salt and calcitriol on the gut-kidney immune system and the composition of the intestinal flora. We demonstrate that consumption of a high-salt diet results in hypertension and inflammation in the colon and kidney and alteration of gut microbiota composition and function. High-salt diet-induced hypertension was found to be associated with seven microbial taxa and mainly associated with reduced production of the protective short-chain fatty acid butyrate. Calcitriol can reduce colon and kidney inflammation, and there are gene expression changes consistent with restored intestinal barrier function. The protective effect of calcitriol may be mediated indirectly by immunological properties. Additionally, the molecular pathways of the gut microbiota-mediated blood pressure regulation may be related to circadian rhythm signals, which needs to be further investigated. An innovative association analysis of the microbiota may be a key strategy to understanding the association between gene patterns and host.
Collapse
Affiliation(s)
- Ruifeng Ding
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Zilong Xiao
- Department of Cardiology, Zhongshan Hospital of Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
| | - Yufeng Jiang
- Department of Nephrology, 66329Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China.,Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai 201203, China
| | - Yi Yang
- Shanghai Cinoasia Institute, Shanghai 200438, China
| | - Yang Ji
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xunxia Bao
- Shanghai Cinoasia Institute, Shanghai 200438, China
| | - Kaichen Xing
- Shanghai Cinoasia Institute, Shanghai 200438, China
| | - Xinli Zhou
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Sibo Zhu
- School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
26
|
Torres-Pinzon DL, Ralph DL, Veiras LC, McDonough AA. Sex-specific adaptations to high-salt diet preserve electrolyte homeostasis with distinct sodium transporter profiles. Am J Physiol Cell Physiol 2021; 321:C897-C909. [PMID: 34613843 PMCID: PMC8616593 DOI: 10.1152/ajpcell.00282.2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023]
Abstract
Kidneys continuously filter an enormous amount of sodium and adapt kidney Na+ reabsorption to match Na+ intake to maintain circulatory volume and electrolyte homeostasis. Males (M) respond to high-salt (HS) diet by translocating proximal tubule Na+/H+ exchanger isoform 3 (NHE3) to the base of the microvilli, reducing activated forms of the distal NaCl cotransporter (NCC) and epithelial Na+ channel (ENaC). Males (M) and females (F) on normal-salt (NS) diet present sex-specific profiles of "transporters" (cotransporters, channels, pumps, and claudins) along the nephron, e.g., F exhibit 40% lower NHE3 and 200% higher NCC abundance than M. We tested the hypothesis that adaptations to HS diet along the nephron will, likewise, exhibit sexual dimorphisms. C57BL/6J mice were fed for 15 days with 4% NaCl diet (HS) versus 0.26% NaCl diet (NS). On HS, M and F exhibited normal plasma [Na+] and [K+], similar urine volume, Na+, K+, and osmolal excretion rates normalized to body weight. In F, like M, HS lowered abundance of distal NCC, phosphorylated NCC, and cleaved (activated) forms of ENaC. The adaptations associated with achieving electrolyte homeostasis exhibit sex-dependent and independent mechanisms. Sex differences in baseline "transporters" abundance persist during HS diet, yet the fold changes during HS diet (normalized to NS) are similar along the distal nephron and collecting duct. Sex-dependent differences observed along the proximal tubule during HS show that female kidneys adapt differently from patterns reported in males, yet achieve and maintain fluid and electrolyte homeostasis.
Collapse
Affiliation(s)
- Diana L Torres-Pinzon
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Donna L Ralph
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Luciana C Veiras
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Alicia A McDonough
- Department of Physiology and Neuroscience, Keck School of Medicine of University of Southern California, Los Angeles, California
| |
Collapse
|
27
|
Kawarazaki W, Fujita T. Kidney and epigenetic mechanisms of salt-sensitive hypertension. Nat Rev Nephrol 2021; 17:350-363. [PMID: 33627838 DOI: 10.1038/s41581-021-00399-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
Dietary salt intake increases blood pressure (BP) but the salt sensitivity of BP differs between individuals. The interplay of ageing, genetics and environmental factors, including malnutrition and stress, contributes to BP salt sensitivity. In adults, obesity is often associated with salt-sensitive hypertension. The children of women who experience malnutrition during pregnancy are at increased risk of developing obesity, diabetes and salt-sensitive hypertension as adults. Similarly, the offspring of mice that are fed a low-protein diet during pregnancy develop salt-sensitive hypertension in association with aberrant DNA methylation of the gene encoding type 1A angiotensin II receptor (AT1AR) in the hypothalamus, leading to upregulation of hypothalamic AT1AR and renal sympathetic overactivity. Ageing is also associated with salt-sensitive hypertension. In aged mice, promoter methylation leads to reduced kidney production of the anti-ageing factor Klotho and a decrease in circulating soluble Klotho. In the setting of Klotho deficiency, salt-induced activation of the vascular Wnt5a-RhoA pathway leads to ageing-associated salt-sensitive hypertension, potentially as a result of reduced renal blood flow and increased peripheral resistance. Thus, kidney mechanisms and aberrant DNA methylation of certain genes are involved in the development of salt-sensitive hypertension during fetal development and old age. Three distinct paradigms of epigenetic memory operate on different timescales in prenatal malnutrition, obesity and ageing.
Collapse
Affiliation(s)
- Wakako Kawarazaki
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan. .,School of Medicine, Shinshu University, Matsumoto, Japan. .,Research Center for Social Systems, Shinshu University, Matsumoto, Japan.
| |
Collapse
|
28
|
Kamiar A, Yousefi K, Dunkley JC, Webster KA, Shehadeh LA. β 2-Adrenergic receptor agonism as a therapeutic strategy for kidney disease. Am J Physiol Regul Integr Comp Physiol 2021; 320:R575-R587. [PMID: 33565369 DOI: 10.1152/ajpregu.00287.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Approximately 14% of the general population suffer from chronic kidney disease that can lead to acute kidney injury (AKI), a condition with up to 50% mortality for which there is no effective treatment. Hypertension, diabetes, and cardiovascular disease are the main comorbidities, and more than 660,000 Americans have kidney failure. β2-Adrenergic receptors (β2ARs) have been extensively studied in association with lung and cardiovascular disease, but with limited scope in kidney and renal diseases. β2ARs are expressed in multiple parts of the kidney including proximal and distal convoluted tubules, glomeruli, and podocytes. Classical and noncanonical β2AR signaling pathways interface with other intracellular mechanisms in the kidney to regulate important cellular functions including renal blood flow, electrolyte balance and salt handling, and tubular function that in turn exert control over critical physiology and pathology such as blood pressure and inflammatory responses. Nephroprotection through activation of β2ARs has surfaced as a promising field of investigation; however, there is limited data on the pharmacology and potential side effects of renal β2AR modulation. Here, we provide updates on some of the major areas of preclinical kidney research involving β2AR signaling that have advanced to describe molecular pathways and identify potential drug targets some of which are currently under clinical development for the treatment of kidney-related diseases.
Collapse
Affiliation(s)
- Ali Kamiar
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Keyvan Yousefi
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Julian C Dunkley
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Keith A Webster
- Vascular Biology Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Division of Cardiology, Department of Medicine, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.,Peggy and Harold Katz Family Drug Discovery Center, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| |
Collapse
|
29
|
Tiyasatkulkovit W, Aksornthong S, Adulyaritthikul P, Upanan P, Wongdee K, Aeimlapa R, Teerapornpuntakit J, Rojviriya C, Panupinthu N, Charoenphandhu N. Excessive salt consumption causes systemic calcium mishandling and worsens microarchitecture and strength of long bones in rats. Sci Rep 2021; 11:1850. [PMID: 33473159 PMCID: PMC7817681 DOI: 10.1038/s41598-021-81413-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Excessive salt intake has been associated with the development of non-communicable diseases, including hypertension with several cardiovascular consequences. Although the detrimental effects of high salt on the skeleton have been reported, longitudinal assessment of calcium balance together with changes in bone microarchitecture and strength under salt loading has not been fully demonstrated. To address these unanswered issues, male Sprague-Dawley rats were fed normal salt diet (NSD; 0.8% NaCl) or high salt diet (HSD; 8% NaCl) for 5 months. Elevation of blood pressure, cardiac hypertrophy and glomerular deterioration were observed in HSD, thus validating the model. The balance studies were performed to monitor calcium input and output upon HSD challenge. The HSD-induced increase in calcium losses in urine and feces together with reduced fractional calcium absorption led to a decrease in calcium retention. With these calcium imbalances, we therefore examined microstructural changes of long bones of the hind limbs. Using the synchrotron radiation x-ray tomographic microscopy, we showed that trabecular structure of tibia and femur of HSD displayed a marked increase in porosity. Consistently, the volumetric micro-computed tomography also demonstrated a significant decrease in trabecular bone mineral density with expansion of endosteal perimeter in the tibia. Interestingly, bone histomorphometric analyses indicated that salt loading caused an increase in osteoclast number together with decreases in osteoblast number and osteoid volume. This uncoupling process of bone remodeling in HSD might underlie an accelerated bone loss and bone structural changes. In conclusion, long-term excessive salt consumption leads to impairment of skeletal mass and integrity possibly through negative calcium balance.
Collapse
Affiliation(s)
- Wacharaporn Tiyasatkulkovit
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.7922.e0000 0001 0244 7875Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Sirion Aksornthong
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Punyanuch Adulyaritthikul
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Pornpailin Upanan
- grid.411825.b0000 0000 9482 780XFaculty of Allied Health Sciences, Burapha University, Chonburi, 20131 Thailand
| | - Kannikar Wongdee
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.411825.b0000 0000 9482 780XFaculty of Allied Health Sciences, Burapha University, Chonburi, 20131 Thailand
| | - Ratchaneevan Aeimlapa
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Jarinthorn Teerapornpuntakit
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.412029.c0000 0000 9211 2704Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000 Thailand
| | - Catleya Rojviriya
- grid.472685.aSynchrotron Light Research Institute (Public Organization), Nakhon Ratchasima, 30000 Thailand
| | - Nattapon Panupinthu
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand
| | - Narattaphol Charoenphandhu
- grid.10223.320000 0004 1937 0490Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400 Thailand ,grid.10223.320000 0004 1937 0490Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170 Thailand ,The Academy of Science, The Royal Society of Thailand, Dusit, Bangkok, 10300 Thailand
| |
Collapse
|
30
|
Preston RA, Afshartous D, Caizapanta EV, Materson BJ, Rodco R, Alonso E, Alonso AB. Thiazide-Sensitive NCC (Sodium-Chloride Cotransporter) in Human Metabolic Syndrome: Sodium Sensitivity and Potassium-Induced Natriuresis. Hypertension 2021; 77:447-460. [PMID: 33390050 DOI: 10.1161/hypertensionaha.120.15933] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The thiazide-sensitive sodium-chloride cotransporter (NCC;SLC12A3) is central to sodium and blood pressure regulation. Metabolic syndrome induces NCC upregulation generating sodium-sensitive hypertension in experimental animal models. We tested the role of NCC in sodium sensitivity in hypertensive humans with metabolic syndrome. Conversely, oral potassium induces NCC downregulation producing potassium-induced natriuresis. We determined the time course and magnitude of potassium-induced natriuresis compared with the natriuresis following hydrochlorothiazide (HCTZ) as a reference standard. We studied 19 obese hypertensive humans with metabolic syndrome during 13-day inpatient confinement. We determined sodium sensitivity by change in 24-hour mean systolic pressure by automated monitor from days 5 (low sodium) to 10 (high sodium). We determined NCC activity by standard 50 mg HCTZ sensitivity test (day 11). We determined potassium-induced natriuresis following 35 mmol KCl (day 13). We determined (1) whether NCC activity was greater in sodium-sensitive versus sodium-resistant participants and correlated with sodium sensitivity and (2) time course and magnitude of potassium-induced natriuresis following 35 mmol KCl directly compared with 50 mg HCTZ. NCC activity was not greater in sodium-sensitive versus sodium-resistant humans and did not correlate with sodium sensitivity. Thirty-five-millimoles KCl produced a rapid natriuresis approximately half that of 50 mg HCTZ with a greater kaliuresis. Our investigation tested a key hypothesis regarding NCC activity in human hypertension and characterized potassium-induced natriuresis following 35 mmol KCl compared with 50 mg HCTZ. In obese hypertensive adults with metabolic syndrome ingesting a high-sodium diet, 35 mmol KCl had a net natriuretic effect approximately half that of 50 mg HCTZ.
Collapse
Affiliation(s)
- Richard A Preston
- From the Clinical Pharmacology Research Unit, Division of Clinical Pharmacology, Department of Medicine, Miller School of Medicine, University of Miami, FL (R.A.P., D.A., E.V.C., B.J.M., R.R., E.A., A.B.A.).,University of Miami Clinical and Translational Science Institutes, FL (R.A.P.).,Peggy and Harold Katz Family Drug Discovery Center, Miami, FL (R.A.P.)
| | - David Afshartous
- From the Clinical Pharmacology Research Unit, Division of Clinical Pharmacology, Department of Medicine, Miller School of Medicine, University of Miami, FL (R.A.P., D.A., E.V.C., B.J.M., R.R., E.A., A.B.A.)
| | - Evelyn V Caizapanta
- From the Clinical Pharmacology Research Unit, Division of Clinical Pharmacology, Department of Medicine, Miller School of Medicine, University of Miami, FL (R.A.P., D.A., E.V.C., B.J.M., R.R., E.A., A.B.A.)
| | - Barry J Materson
- From the Clinical Pharmacology Research Unit, Division of Clinical Pharmacology, Department of Medicine, Miller School of Medicine, University of Miami, FL (R.A.P., D.A., E.V.C., B.J.M., R.R., E.A., A.B.A.)
| | - Rolando Rodco
- From the Clinical Pharmacology Research Unit, Division of Clinical Pharmacology, Department of Medicine, Miller School of Medicine, University of Miami, FL (R.A.P., D.A., E.V.C., B.J.M., R.R., E.A., A.B.A.)
| | - Eileen Alonso
- From the Clinical Pharmacology Research Unit, Division of Clinical Pharmacology, Department of Medicine, Miller School of Medicine, University of Miami, FL (R.A.P., D.A., E.V.C., B.J.M., R.R., E.A., A.B.A.)
| | - Alberto B Alonso
- From the Clinical Pharmacology Research Unit, Division of Clinical Pharmacology, Department of Medicine, Miller School of Medicine, University of Miami, FL (R.A.P., D.A., E.V.C., B.J.M., R.R., E.A., A.B.A.)
| |
Collapse
|
31
|
Hall JE, Mouton AJ, da Silva AA, Omoto ACM, Wang Z, Li X, do Carmo JM. Obesity, kidney dysfunction, and inflammation: interactions in hypertension. Cardiovasc Res 2020; 117:1859-1876. [PMID: 33258945 DOI: 10.1093/cvr/cvaa336] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/01/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
Obesity contributes 65-75% of the risk for human primary (essential) hypertension (HT) which is a major driver of cardiovascular and kidney diseases. Kidney dysfunction, associated with increased renal sodium reabsorption and compensatory glomerular hyperfiltration, plays a key role in initiating obesity-HT and target organ injury. Mediators of kidney dysfunction and increased blood pressure include (i) elevated renal sympathetic nerve activity (RSNA); (ii) increased antinatriuretic hormones such as angiotensin II and aldosterone; (iii) relative deficiency of natriuretic hormones; (iv) renal compression by fat in and around the kidneys; and (v) activation of innate and adaptive immune cells that invade tissues throughout the body, producing inflammatory cytokines/chemokines that contribute to vascular and target organ injury, and exacerbate HT. These neurohormonal, renal, and inflammatory mechanisms of obesity-HT are interdependent. For example, excess adiposity increases the adipocyte-derived cytokine leptin which increases RSNA by stimulating the central nervous system proopiomelanocortin-melanocortin 4 receptor pathway. Excess visceral, perirenal and renal sinus fat compress the kidneys which, along with increased RSNA, contribute to renin-angiotensin-aldosterone system activation, although obesity may also activate mineralocorticoid receptors independent of aldosterone. Prolonged obesity, HT, metabolic abnormalities, and inflammation cause progressive renal injury, making HT more resistant to therapy and often requiring multiple antihypertensive drugs and concurrent treatment of dyslipidaemia, insulin resistance, diabetes, and inflammation. More effective anti-obesity drugs are needed to prevent the cascade of cardiorenal, metabolic, and immune disorders that threaten to overwhelm health care systems as obesity prevalence continues to increase.
Collapse
Affiliation(s)
- John E Hall
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Clinical and Translational Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA
| | - Alan J Mouton
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA
| | - Alexandre A da Silva
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA
| | - Ana C M Omoto
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA
| | - Zhen Wang
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA
| | - Xuan Li
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA
| | - Jussara M do Carmo
- Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 30216-4505, USA
| |
Collapse
|
32
|
Hu J, Chu C, Shi T, Yan Y, Mu J. Effects of salt intervention on serum levels of Klotho influenced by salt sensitivity. J Clin Hypertens (Greenwich) 2020; 22:2051-2058. [PMID: 33164306 DOI: 10.1111/jch.14044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Jia‐Wen Hu
- Department of Cardiovascular Surgery First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
| | - Chao Chu
- Department of Cardiology First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
- Key Laboratory of Molecular Cardiology of Shaanxi Province Xi’an China
| | - Tao Shi
- Department of Cardiovascular Surgery First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
| | - Yang Yan
- Department of Cardiovascular Surgery First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
| | - Jian‐Jun Mu
- Department of Cardiology First Affiliated Hospital of Medical School Xi’an Jiaotong University Xi’an China
- Key Laboratory of Molecular Cardiology of Shaanxi Province Xi’an China
| |
Collapse
|
33
|
Molla MD, Akalu Y, Geto Z, Dagnew B, Ayelign B, Shibabaw T. Role of Caspase-1 in the Pathogenesis of Inflammatory-Associated Chronic Noncommunicable Diseases. J Inflamm Res 2020; 13:749-764. [PMID: 33116753 PMCID: PMC7585796 DOI: 10.2147/jir.s277457] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Caspase-1 is the first and extensively studied inflammatory caspase that is activated through inflammasome assembly. Inflammasome is a cytosolic formation of multiprotein complex that aimed to start inflammatory response against infections or cellular damages. The process leads to an auto-activation of caspase-1 and consequent maturation of caspase-1 target molecules such as interleukin (IL)-1β and IL-18. Recently, the role of caspase-1 and inflammasome in inflammatory-induced noncommunicable diseases (NCDs) like obesity, diabetes mellitus (DM), cardiovascular diseases (CVDs), cancers and chronic respiratory diseases have widely studied. However, their reports are distinct and even they have reported contrasting role of caspase-1 in the development and progression of NCDs. A few studies have reported that caspase-1/inflammasome assembley has a protective role in the initiation and progression of these diseases through the activation of the noncanonical caspase-1 target substrates like gasdermin-D and regulation of immune cells. Conversely, others have revealed that caspase-1 has a direct/indirect effect in the development and progression of several NCDs. Therefore, in this review, we systematically summarized the role of caspase-1 in the development and progression of NCDs, especially in obesity, DM, CVDs and cancers.
Collapse
Affiliation(s)
- Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yonas Akalu
- Department of Human Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Zeleke Geto
- Department of Biomedical Sciences, School of Medicine, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Baye Dagnew
- Department of Human Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
34
|
Ito K. Review of the health benefits of habitual consumption of miso soup: focus on the effects on sympathetic nerve activity, blood pressure, and heart rate. Environ Health Prev Med 2020; 25:45. [PMID: 32867671 PMCID: PMC7461326 DOI: 10.1186/s12199-020-00883-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023] Open
Abstract
High salt intake increases blood pressure, and dietary salt intake has been clearly demonstrated to be associated with hypertension incidence. Japanese people consume higher amounts of salt than Westerners. It has been reported that miso soup was one of the major sources of daily salt intake in Japanese people. Adding salt is indispensable to make miso, and therefore, in some cases, refraining from miso soup is recommended to reduce dietary salt intake. However, recent studies using salt-sensitive hypertensive models have revealed that miso lessens the effects of salt on blood pressure. In other word, the intake of miso dose not increase the blood pressure compared to the equivalent intake of salt. In addition, many clinical observational studies have demonstrated the absence of a relationship between the frequency of miso soup intake and blood pressure levels or hypertension incidence. The mechanism of this phenomenon seen in the subjects with miso soup intake has not been fully elucidated yet. However, in basic studies, it was found that the ingredients of miso attenuate sympathetic nerve activity, resulting in lowered blood pressure and heart rate. Therefore, this review focused on the differences between the effects of miso intake and those of the equivalent salt intake on sympathetic nerve activity, blood pressure, and heart rate.
Collapse
Affiliation(s)
- Koji Ito
- Department of Clinical Laboratory, Japan Community Healthcare Organization, Kyushu Hospital, 1-8-1, Kishinoura, Yahatanishi-ku, Kitakyushu, 806-8501, Japan.
| |
Collapse
|
35
|
Wu J, Nie J, Wang Y, Zhang Y, Wu D. Relationship between saline infusion and blood pressure variability in non-critically patients with hypertension: A retrospective study. Medicine (Baltimore) 2020; 99:e21468. [PMID: 32871869 PMCID: PMC7458164 DOI: 10.1097/md.0000000000021468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/23/2020] [Accepted: 06/25/2020] [Indexed: 12/29/2022] Open
Abstract
Saline is a commonly used intravenous solvent, however, its excessive infusion may increase drug-induced sodium intake. To investigate the effects of saline infusion on blood pressure variability (BPV) in patients with hypertension, a retrospective study was performed in 1010 patients with hypertension. The patients who received saline infusion before surgery for continuous 3 to 5 days were divided into 2 groups according to the saline infusion volume during the hospitalization, which are >500 mL per day group and <500 mL per day group. The overall incidence of abnormal BPV was 11.58%. As for the incidence of abnormal BPV in the <500 mL per day group with 698 patients was 9.17%, while that in the >500 mL per day group with 312 patients was as high as 16.99%. Additionally, >500 mL of daily saline infusion for continuous 3 to 5 days (P for trend = .004, odds ratio [OR] = 1.911, 95% confidence interval [CI] for OR 1.226-2.977), medical history of diabetes mellitus (P < .001, OR = 4.856, 95% CI for OR 3.118-7.563) and cardiovascular diseases (P < .001, OR = 2.498, 95% CI for OR 1.549-4.029) may be risk factors of abnormal BPV; while anti-hypertensive therapy with diuretics (P < .001, OR = 0.055, 95% CI for OR 0.024-0.125) may be the protective factor. Our study suggests that >500 mL of daily saline infusion for continuous 3 to 5 days may have disadvantages in the blood pressure control for hypertensive patients, especially for the patients with diabetes mellitus and cardiovascular diseases.
Collapse
|
36
|
Woodman AG, Mah R, Keddie DL, Noble RMN, Holody CD, Panahi S, Gragasin FS, Lemieux H, Bourque SL. Perinatal iron deficiency and a high salt diet cause long-term kidney mitochondrial dysfunction and oxidative stress. Cardiovasc Res 2020; 116:183-192. [PMID: 30715197 DOI: 10.1093/cvr/cvz029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/08/2019] [Accepted: 01/29/2019] [Indexed: 12/15/2022] Open
Abstract
AIMS Perinatal iron deficiency (ID) alters developmental trajectories of offspring, predisposing them to cardiovascular dysfunction in later life. The mechanisms underlying this long-term programming of renal function have not been defined. We hypothesized perinatal ID causes hypertension and alters kidney metabolic function and morphology in a sex-dependent manner in adult offspring. Furthermore, we hypothesized these effects are exacerbated by chronic consumption of a high salt diet. METHODS AND RESULTS Pregnant Sprague Dawley rats were fed either an iron-restricted or replete diet prior to and throughout pregnancy. Adult offspring were fed normal or high salt diets for 6 weeks prior to experimentation at 6 months of age. Blood pressure (BP) was assessed via indwelling catheters in anaesthetized offspring; kidney mitochondrial function was assessed via high-resolution respirometry; reactive oxygen species and nitric oxide were quantified via fluorescence microscopy. Adult males, but not females, exhibited increased systolic BP due to ID (P = 0.01) and high salt intake (P = 0.02). In males, but not in females, medullary mitochondrial content was increased by high salt (P = 0.003), while succinate-dependent respiration was reduced by ID (P < 0.05). The combination of perinatal ID and high salt reduced complex IV activity in the cortex of males (P = 0.01). Perinatal ID increased cytosolic superoxide generation (P < 0.001) concomitant with reduced nitric oxide bioavailability (P < 0.001) in male offspring, while high salt increased mitochondrial superoxide in the medulla (P = 0.04) and cytosolic superoxide within the cortex (P = 0.01). Male offspring exhibited glomerular basement membrane thickening (P < 0.05), increased collagen deposition (P < 0.05), and glomerular hypertrophy (interaction, P = 0.02) due to both perinatal ID and high salt. Female offspring exhibited no alterations in mitochondrial function or morphology due to either high salt or ID. CONCLUSION Perinatal ID causes long-term sex-dependent alterations in renal metabolic function and morphology, potentially contributing to hypertension and increased cardiovascular disease risk.
Collapse
Affiliation(s)
- Andrew G Woodman
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Richard Mah
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Danae L Keddie
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Ronan M N Noble
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Claudia D Holody
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Sareh Panahi
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| | - Ferrante S Gragasin
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| | - Helene Lemieux
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Medicine, University of Alberta, Edmonton, Canada.,Faculty Saint-Jean, University of Alberta, Edmonton, Canada
| | - Stephane L Bourque
- Department of Pharmacology, University of Alberta, Edmonton, Canada.,Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada.,Department of Pediatrics, University of Alberta, Edmonton, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
37
|
Kishimoto T, Kataoka T, Yamamoto Y, Asano G, Fukamoto A, Hotta Y, Maeda Y, Takahashi M, Kanayama HO, Kimura K. High Salt Intake Impairs Erectile Function in Salt-Sensitive Rats Through Mineralocorticoid Receptor Pathway Beyond Its Effect on Blood Pressure. J Sex Med 2020; 17:1280-1287. [DOI: 10.1016/j.jsxm.2020.04.384] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/30/2020] [Accepted: 04/23/2020] [Indexed: 12/15/2022]
|
38
|
Hirohama D, Kawarazaki W, Nishimoto M, Ayuzawa N, Marumo T, Shibata S, Fujita T. PGI 2 Analog Attenuates Salt-Induced Renal Injury through the Inhibition of Inflammation and Rac1-MR Activation. Int J Mol Sci 2020; 21:ijms21124433. [PMID: 32580367 PMCID: PMC7353033 DOI: 10.3390/ijms21124433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/18/2022] Open
Abstract
Renal inflammation is known to be involved in salt-induced renal damage, leading to end-stage renal disease. This study aims to evaluate the role of inflammation in anti-inflammatory and renoprotective effects of beraprost sodium (BPS), a prostaglandin I2 (PGI2) analog, in Dahl salt-sensitive (DS) rats. Five-week-old male DS rats were fed a normal-salt diet (0.5% NaCl), a high-salt diet (8% NaCl), or a high-salt diet plus BPS treatment for 3 weeks. BPS treatment could inhibit marked proteinuria and renal injury in salt-loaded DS rats with elevated blood pressure, accompanied by renal inflammation suppression. Notably, high salt increased renal expression of active Rac1, followed by increased Sgk1 expressions, a downstream molecule of mineralocorticoid receptor (MR) signal, indicating salt-induced activation of Rac1-MR pathway. However, BPS administration inhibited salt-induced Rac1-MR activation as well as renal inflammation and damage, suggesting that Rac1-MR pathway is involved in anti-inflammatory and renoprotective effects of PGI2. Based upon Rac1 activated by inflammation, moreover, BPS inhibited salt-induced activation of Rac1-MR pathway by renal inflammation suppression, resulting in the attenuation of renal damage in salt-loaded DS rats. Thus, BPS is efficacious for the treatment of salt-induced renal injury.
Collapse
Affiliation(s)
- Daigoro Hirohama
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (W.K.); (M.N.); (N.A.); (T.M.); (S.S.); (T.F.)
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan
- Correspondence: ; Tel.: +81-3-5452-5057
| | - Wakako Kawarazaki
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (W.K.); (M.N.); (N.A.); (T.M.); (S.S.); (T.F.)
| | - Mitsuhiro Nishimoto
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (W.K.); (M.N.); (N.A.); (T.M.); (S.S.); (T.F.)
- Department of Internal Medicine, International University of Health and Welfare Mita Hospital, Tokyo 108-8329, Japan
| | - Nobuhiro Ayuzawa
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (W.K.); (M.N.); (N.A.); (T.M.); (S.S.); (T.F.)
| | - Takeshi Marumo
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (W.K.); (M.N.); (N.A.); (T.M.); (S.S.); (T.F.)
- Center for Basic Medical Research at Narita Campus, International University of Health and Welfare, Chiba 286-8686, Japan
| | - Shigeru Shibata
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (W.K.); (M.N.); (N.A.); (T.M.); (S.S.); (T.F.)
- Division of Nephrology, Department of Internal Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo 153-8904, Japan; (W.K.); (M.N.); (N.A.); (T.M.); (S.S.); (T.F.)
- Shinshu University School of Medicine and Research Center for Social Systems, Nagano 389-0111, Japan
| |
Collapse
|
39
|
Rao X, Asico LD, Zanos P, Mahabeleshwar GH, Singh Gangwar R, Xia C, Duan L, Cisse YM, Rengasamy P, Jose PA, Gould TD, Nelson R, Biswal S, Chen LC, Zhong J, Rajagopalan S. Alpha2B-Adrenergic Receptor Overexpression in the Brain Potentiate Air Pollution-induced Behavior and Blood Pressure Changes. Toxicol Sci 2020; 169:95-107. [PMID: 30812033 DOI: 10.1093/toxsci/kfz025] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Fine ambient particulate matter (PM2.5) is able to induce sympathetic activation and inflammation in the brain. However, direct evidence demonstrating an essential role of sympathetic activation in PM2.5-associated disease progression is lacking. We assess the contribution of α2B-adrenergic receptor (Adra2b) in air pollution-associated hypertension and behavioral changes in this study. Wild-type mice and Adra2b-transgenic mice overexpressing Adra2b in the brain (Adra2bTg) were exposed to concentrated PM2.5 or filtered air for 3 months via a versatile aerosol concentrator exposure system. Mice were fed with a high salt diet (4.0% NaCl) for 1 week at week 11 of exposure to induce blood pressure elevation. Intra-arterial blood pressure was monitored by radio-telemetry and behavior changes were assessed by open field, light-dark, and prepulse inhibition tests. PM2.5 exposure increased Adra2b in the brain of wild-type mice. Adra2b overexpression enhanced the anxiety-like behavior and high salt diet-induced blood pressure elevation in response to air pollution but not filtered air exposure. Adra2b overexpression induced upregulation of inflammatory genes such as TLR2, TLR4, and IL-6 in the brain exposed to PM2.5. In addition, there were increased frequencies of activated effector T cells and increased expression of oxidative stress-related genes, such as SOD1, NQO1, Nrf2, and Gclm in Adra2bTg mice compared with wild-type mice. Our results provide new evidence of distinct behavioral changes consistent with anxiety and blood pressure elevation in response to high salt intake and air pollution exposure, highlighting the importance of centrally expressed Adra2b in the vulnerability to air pollution exposure.
Collapse
Affiliation(s)
- Xiaoquan Rao
- Oregon Institute of Occupational Health Science, Oregon Health & Science University, Portland, Oregon.,Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Laureano D Asico
- Division of Renal Diseases & Hypertension, The George Washington University, Washington, District of Columbia
| | - Panos Zanos
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | | | | | - Chang Xia
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Lihua Duan
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio
| | | | - Palanivel Rengasamy
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, The George Washington University, Washington, District of Columbia
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Randy Nelson
- Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Shyam Biswal
- Department of Environmental Health Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Lung-Chi Chen
- Department of Environmental Medicine, New York University, Tuxedo, New York
| | - Jixin Zhong
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
40
|
Yan X, Jin J, Su X, Yin X, Gao J, Wang X, Zhang S, Bu P, Wang M, Zhang Y, Wang Z, Zhang Q. Intestinal Flora Modulates Blood Pressure by Regulating the Synthesis of Intestinal-Derived Corticosterone in High Salt-Induced Hypertension. Circ Res 2020; 126:839-853. [DOI: 10.1161/circresaha.119.316394] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rationale:
High-salt diet is one of the most important risk factors for hypertension. Intestinal flora has been reported to be associated with high salt–induced hypertension (hSIH). However, the detailed roles of intestinal flora in hSIH pathogenesis have not yet been fully elucidated.
Objective:
To reveal the roles and mechanisms of intestinal flora in hSIH development.
Methods and Results:
The abovementioned issues were investigated using various techniques including 16S rRNA gene sequencing, untargeted metabolomics, selective bacterial culture, and fecal microbiota transplantation. We found that high-salt diet induced hypertension in Wistar rats. The fecal microbiota of healthy rats could dramatically lower blood pressure (BP) of hypertensive rats, whereas the fecal microbiota of hSIH rats had opposite effects. The composition, metabolism, and interrelationship of intestinal flora in hSIH rats were considerably reshaped, including the increased corticosterone level and reduced
Bacteroides
and arachidonic acid levels, which tightly correlated with BP. The serum corticosterone level was also significantly increased in rats with hSIH. Furthermore, the above abnormalities were confirmed in patients with hypertension. The intestinal
Bacteroides fragilis
could inhibit the production of intestinal-derived corticosterone induced by high-salt diet through its metabolite arachidonic acid.
Conclusions:
hSIH could be transferred by fecal microbiota transplantation, indicating the pivotal roles of intestinal flora in hSIH development. High-salt diet reduced the levels of
B fragilis
and arachidonic acid in the intestine, which increased intestinal-derived corticosterone production and corticosterone levels in serum and intestine, thereby promoting BP elevation. This study revealed a novel mechanism different from inflammation/immunity by which intestinal flora regulated BP, namely intestinal flora could modulate BP by affecting steroid hormone levels. These findings enriched the understanding of the function of intestinal flora and its effects on hypertension.
Collapse
Affiliation(s)
- Xuefang Yan
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Jiajia Jin
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Xinhuan Su
- Division of Endocrinology and Metabolism (X.S., Z.W.), Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Division of Geriatrics (X.S., Z.W.), Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Xianlun Yin
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Jing Gao
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Xiaowei Wang
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Shucui Zhang
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Peili Bu
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Mansen Wang
- Medical Data Research Center, Providence Health & Services, Portland, OR (M.W.)
| | - Yun Zhang
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| | - Zhe Wang
- Division of Endocrinology and Metabolism (X.S., Z.W.), Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
- Division of Geriatrics (X.S., Z.W.), Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Qunye Zhang
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan (X. Yan, J.J., X. Yin, J.G., X.W., S.Z., P.B., Y.Z., Q.Z.)
| |
Collapse
|
41
|
Wan N, Fujisawa Y, Kobara H, Masaki T, Nakano D, Rahman A, Nishiyama A. Effects of an SGLT2 inhibitor on the salt sensitivity of blood pressure and sympathetic nerve activity in a nondiabetic rat model of chronic kidney disease. Hypertens Res 2020; 43:492-499. [PMID: 32060381 DOI: 10.1038/s41440-020-0410-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/09/2019] [Accepted: 12/23/2019] [Indexed: 12/20/2022]
Abstract
The glucose-lowering effect of sodium-glucose cotransporter 2 (SGLT2) inhibitors is reduced in patients with diabetes who have chronic kidney disease (CKD). In the present study, we examined the effect of an SGLT2 inhibitor on the salt sensitivity of blood pressure (BP), circadian rhythm of BP, and sympathetic nerve activity (SNA) in nondiabetic CKD rats. Uninephrectomized Wistar rats were treated with adenine (200 mg/kg/day) for 14 days. After stabilization with a normal-salt diet (NSD, 0.3% NaCl), a high-salt diet (HSD, 8% NaCl) was administered. Mean arterial pressure (MAP) was continuously monitored using a telemetry system. We also analyzed the low frequency (LF) of systolic arterial pressure (SAP), which reflects SNA. In adenine-induced CKD rats, HSD consumption for 5 days significantly increased the mean MAP from 106 ± 2 to 148 ± 3 mmHg. However, MAP was decreased to 96 ± 3 mmHg within 24 h after switching back to a NSD (n = 7). Treatment with an SGLT2 inhibitor, luseogliflozin (10 mg/kg/day, p.o., n = 7), significantly attenuated the HSD-induced elevation of MAP, which was associated with a reduction in LF of SAP. These data suggest that treatment with an SGLT2 inhibitor attenuates the salt sensitivity of BP, which is associated with SNA inhibition in nondiabetic CKD rats.
Collapse
Affiliation(s)
- Ningning Wan
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yoshihide Fujisawa
- Life Science Research Center, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Asadur Rahman
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| |
Collapse
|
42
|
Hall JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity, kidney dysfunction and hypertension: mechanistic links. Nat Rev Nephrol 2020; 15:367-385. [PMID: 31015582 DOI: 10.1038/s41581-019-0145-4] [Citation(s) in RCA: 327] [Impact Index Per Article: 65.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Excessive adiposity raises blood pressure and accounts for 65-75% of primary hypertension, which is a major driver of cardiovascular and kidney diseases. In obesity, abnormal kidney function and associated increases in tubular sodium reabsorption initiate hypertension, which is often mild before the development of target organ injury. Factors that contribute to increased sodium reabsorption in obesity include kidney compression by visceral, perirenal and renal sinus fat; increased renal sympathetic nerve activity (RSNA); increased levels of anti-natriuretic hormones, such as angiotensin II and aldosterone; and adipokines, particularly leptin. The renal and neurohormonal pathways of obesity and hypertension are intertwined. For example, leptin increases RSNA by stimulating the central nervous system proopiomelanocortin-melanocortin 4 receptor pathway, and kidney compression and RSNA contribute to renin-angiotensin-aldosterone system activation. Glucocorticoids and/or oxidative stress may also contribute to mineralocorticoid receptor activation in obesity. Prolonged obesity and progressive renal injury often lead to the development of treatment-resistant hypertension. Patient management therefore often requires multiple antihypertensive drugs and concurrent treatment of dyslipidaemia, insulin resistance, diabetes and inflammation. If more effective strategies for the prevention and control of obesity are not developed, cardiorenal, metabolic and other obesity-associated diseases could overwhelm health-care systems in the future.
Collapse
Affiliation(s)
- John E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA. .,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA.
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alexandre A da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Zhen Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA.,Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA.,Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
43
|
Role of Hyperinsulinemia and Insulin Resistance in Hypertension: Metabolic Syndrome Revisited. Can J Cardiol 2020; 36:671-682. [PMID: 32389340 DOI: 10.1016/j.cjca.2020.02.066] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023] Open
Abstract
Hyperinsulinemia and insulin resistance were proposed more than 30 years ago to be important contributors to elevated blood pressure (BP) associated with obesity and the metabolic syndrome, also called syndrome X. Support for this concept initially came from clinical and population studies showing correlations among hyperinsulinemia, insulin resistance, and elevated BP in individuals with metabolic syndrome. Short-term studies in experimental animals and in humans provided additional evidence that hyperinsulinemia may evoke increases in sympathetic nervous system (SNS) activity and renal sodium retention that, if sustained, could increase BP. Although insulin infusions may increase SNS activity and modestly raise BP in rodents, chronic insulin administration does not significantly increase BP in lean or obese insulin-resistant rabbits, dogs, horses, or humans. Multiple studies in humans and experimental animals have also shown that severe insulin resistance and hyperinsulinemia may occur in the absence of elevated BP. These observations question whether insulin resistance and hyperinsulinemia are major factors linking obesity/metabolic syndrome with hypertension. Other mechanisms, such as physical compression of the kidneys, activation of the renin-angiotensin-aldosterone system, hyperleptinemia, stimulation of the brain melanocortin system, and SNS activation, appear to play a more critical role in initiating hypertension in obese subjects with metabolic syndrome. However, the metabolic effects of insulin resistance, including hyperglycemia and dyslipidemia, appear to interact synergistically with increased BP to cause vascular and kidney injury that can exacerbate the hypertension and associated injury to the kidneys and cardiovascular system.
Collapse
|
44
|
GRK2-Mediated Crosstalk Between β-Adrenergic and Angiotensin II Receptors Enhances Adrenocortical Aldosterone Production In Vitro and In Vivo. Int J Mol Sci 2020; 21:ijms21020574. [PMID: 31963151 PMCID: PMC7013621 DOI: 10.3390/ijms21020574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/06/2020] [Accepted: 01/13/2020] [Indexed: 12/13/2022] Open
Abstract
Aldosterone is produced by adrenocortical zona glomerulosa (AZG) cells in response to angiotensin II (AngII) acting through its type I receptors (AT1Rs). AT1R is a G protein-coupled receptor (GPCR) that induces aldosterone via both G proteins and the adapter protein βarrestin1, which binds the receptor following its phosphorylation by GPCR-kinases (GRKs) to initiate G protein-independent signaling. β-adrenergic receptors (ARs) also induce aldosterone production in AZG cells. Herein, we investigated whether GRK2 or GRK5, the two major adrenal GRKs, is involved in the catecholaminergic regulation of AngII-dependent aldosterone production. In human AZG (H295R) cells in vitro, the βAR agonist isoproterenol significantly augmented both AngII-dependent aldosterone secretion and synthesis, as measured by the steroidogenic acute regulatory (StAR) protein and CYP11B2 (aldosterone synthase) mRNA inductions. Importantly, GRK2, but not GRK5, was indispensable for the βAR-mediated enhancement of aldosterone in response to AngII. Specifically, GRK2 inhibition with Cmpd101 abolished isoproterenol’s effects on AngII-induced aldosterone synthesis/secretion, whereas the GRK5 knockout via CRISPR/Cas9 had no effect. It is worth noting that these findings were confirmed in vivo, since rats overexpressing GRK2, but not GRK5, in their adrenals had elevated circulating aldosterone levels compared to the control animals. However, treatment with the β-blocker propranolol prevented hyperaldosteronism in the adrenal GRK2-overexpressing rats. In conclusion, GRK2 mediates a βAR-AT1R signaling crosstalk in the adrenal cortex leading to elevated aldosterone production. This suggests that adrenal GRK2 may be a molecular link connecting the sympathetic nervous and renin-angiotensin systems at the level of the adrenal cortex and that its inhibition might be therapeutically advantageous in hyperaldosteronism-related conditions.
Collapse
|
45
|
Dalmasso C, Leachman JR, Osborn JL, Loria AS. Sensory signals mediating high blood pressure via sympathetic activation: role of adipose afferent reflex. Am J Physiol Regul Integr Comp Physiol 2019; 318:R379-R389. [PMID: 31868518 DOI: 10.1152/ajpregu.00079.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Blood pressure regulation in health and disease involves a balance between afferent and efferent signals from multiple organs and tissues. Although there are numerous reviews focused on the role of sympathetic nerves in different models of hypertension, few have revised the contribution of afferent nerves innervating adipose tissue and their role in the development of obesity-induced hypertension. Both clinical and basic research support the beneficial effects of bilateral renal denervation in lowering blood pressure. However, recent studies revealed that afferent signals from adipose tissue, in an adipose-brain-peripheral pathway, could contribute to the increased sympathetic activation and blood pressure during obesity. This review focuses on the role of adipose tissue afferent reflexes and briefly describes a number of other afferent reflexes modulating blood pressure. A comprehensive understanding of how multiple afferent reflexes contribute to the pathophysiology of essential and/or obesity-induced hypertension may provide significant insights into improving antihypertensive therapeutic approaches.
Collapse
Affiliation(s)
- Carolina Dalmasso
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jacqueline R Leachman
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Jeffrey L Osborn
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
46
|
Agbaraolorunpo FM, Sofola OA, Anigbogu CN, Azinge EC. Angiotensin receptor blockade with Losartan attenuates pressor response to handgrip contraction and enhances natriuresis in salt loaded hypertensive subjects: a quasi-experimental study among Nigerian adults. Pan Afr Med J 2019; 34:188. [PMID: 32180862 PMCID: PMC7060958 DOI: 10.11604/pamj.2019.34.188.18317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/18/2019] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Sympathetic and Renin-Angiotensin-Aldosterone systems play crucial roles in blood pressure response to increased salt intake. This study investigated the effects of angiotensin receptor blocker (ARB) and sympathetic excitation on the responses of blood pressure (BP) and peripheral vascular resistance (PVR) in salt loaded normotensive (NT) and hypertensive (HT) Nigerian subjects. METHODS 16 NT and 14 HT participants, that were age-matched [39.9 ± 1.3 vs 44.1±2.1yrs (P= 0.10)], underwent 5 days each of oral administration of 200mmol NaCl, and 200mmol NaCl + 50mg Losartan, preceded by a baseline control condition. BP and PVR responses to 30% Maximum Voluntary Contraction (MVC) of handgrip (HG) for one minute were determined at baseline, after salt load and after salt + Losartan. Data were presented as Mean ± SEM, and analyzed with two-way ANOVA and paired t-test, with P<0.05 accepted as significant. RESULTS BP and PVR were significantly increased by HG at baseline, after salt load and after salt + Losartan in NT and HT. Salt load augmented the HG-induced SBP (P=0.04) and MABP responses (P=0.02) in HT. While Losartan attenuated the HG- induced Systolic Blood Pressure (SBP) SBP response (P=0.007) and DBP response (P=0.003) in HT and NT respectively after salt + Losartan. HG-induced PVR response was significantly accentuated after salt load in HT (P=0.005), but it was not significant in NT (P=0.38). CONCLUSION The implication of our finding is that angiotensin II receptor blockade possibly attenuates salt-induced sympathetic nerve excitation in black hypertensive patients.
Collapse
Affiliation(s)
| | | | | | - Elaine Chinyelu Azinge
- Department of Clinical Pathology, College of Medicine, University of Lagos, Lagos, Nigeria
| |
Collapse
|
47
|
Pablo JL, Greka A. Charting a TRP to Novel Therapeutic Destinations for Kidney Diseases. Trends Pharmacol Sci 2019; 40:911-918. [PMID: 31704171 DOI: 10.1016/j.tips.2019.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 12/29/2022]
Abstract
Ion channels are critical to kidney function, and their dysregulation leads to several distinct kidney diseases. Of the diversity of ion channels in kidney cells, the transient receptor potential (TRP) superfamily of proteins plays important and varied roles in both maintaining homeostasis as well as in causing disease. Recent work showed that TRPC5 blockers could successfully protect critical components of the kidney filter both in vitro and in vivo, thus revealing TRPC5 as a tractable therapeutic target for focal and segmental glomerulosclerosis (FSGS), a common cause of kidney failure. Human genetics point to three additional TRP channels as plausible therapeutic targets: TRPC6 in FSGS, PKD2 in polycystic kidney disease, and TRPM6 in familial hypomagnesemia with secondary hypocalcemia (HSH). We conclude that targeting TRP channels could pave the way for much needed therapies for kidney diseases.
Collapse
Affiliation(s)
- Juan Lorenzo Pablo
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Anna Greka
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
48
|
Zicha J, Hojná S, Vaňourková Z, Kopkan L, Vaněčková I. Is renal ß-adrenergic-WNK4-NCC pathway important in salt hypertension of Dahl rats? Physiol Res 2019; 68:873-882. [PMID: 31647304 DOI: 10.33549/physiolres.934334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In 2011 Fujita and coworkers proposed that ß-adrenergic stimulation causes decreased serine/threonine-protein kinase WNK4 transcription leading to the activation of Na-Cl cotransporter (NCC) which participates in salt sensitivity and salt hypertension development in rodents. The aim of our study was to investigate whether the above hypothesis is also valid for salt hypertension of Dahl rats, which are characterized by high sympathetic tone and abnormal renal sodium handling. Male 8-week-old salt-sensitive (SS/Jr) and salt-resistant (SR/Jr) Dahl rats were fed either low-salt diet (LS, 0.4 % NaCl) or high-salt diet (HS, 4 % NaCl) for 6 weeks. Half of the animals on either diet were chronically treated with non-selective ß-blocker propranolol (100 mg/kg/day). At the end of the experiment diuresis and sodium excretion were measured prior and after hydrochlorothiazide injection (HCTZ, 10 mg/kg i.p.). Furthermore, blood pressure (BP), heart rate (HR), sympathetic (pentolinium 5 mg/kg i.v.) and NO-dependent (L-NAME 30 mg/kg i.v.) BP components were determined. Chronic HS diet feeding increased BP through sympathoexcitation in SS/Jr but not in SR/Jr rats. Concomitant propranolol treatment did not lower BP in either experimental group. Under the conditions of low salt intake HCTZ increased diuresis, natriuresis and fractional sodium excretion in SR/Jr but not in SS/Jr rats. HS diet feeding attenuated renal response to HCT in SR/Jr rats, whereas no HCTZ effect was observed in SS/Jr rats fed HS diet. Propranolol treatment did not modify diuresis or natriuresis in any experimental group. In conclusions, our present data do not support the idea on the essential importance of renal ß-adrenergic-WNK4-NCC pathway in pathogenesis and/or maintenance of salt hypertension in Dahl rats.
Collapse
Affiliation(s)
- J Zicha
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
49
|
Campbell NRC, He FJ, Tan M, Cappuccio FP, Neal B, Woodward M, Cogswell ME, McLean R, Arcand J, MacGregor G, Whelton P, Jula A, L'Abbe MR, Cobb LK, Lackland DT. The International Consortium for Quality Research on Dietary Sodium/Salt (TRUE) position statement on the use of 24-hour, spot, and short duration (<24 hours) timed urine collections to assess dietary sodium intake. J Clin Hypertens (Greenwich) 2019; 21:700-709. [PMID: 31087778 DOI: 10.1111/jch.13551] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 12/26/2022]
Abstract
The International Consortium for Quality Research on Dietary Sodium/Salt (TRUE) is a coalition of intentional and national health and scientific organizations formed because of concerns low-quality research methods were creating controversy regarding dietary salt reduction. One of the main sources of controversy is believed related to errors in estimating sodium intake with urine studies. The recommendations and positions in this manuscript were generated following a series of systematic reviews and analyses by experts in hypertension, nutrition, statistics, and dietary sodium. To assess the population's current 24-hour dietary sodium ingestion, single complete 24-hour urine samples, collected over a series of days from a representative population sample, were recommended. To accurately estimate usual dietary sodium at the individual level, at least 3 non-consecutive complete 24-hour urine collections obtained over a series of days that reflect the usual short-term variations in dietary pattern were recommended. Multiple 24-hour urine collections over several years were recommended to estimate an individual's usual long-term sodium intake. The role of single spot or short duration timed urine collections in assessing population average sodium intake requires more research. Single or multiple spot or short duration timed urine collections are not recommended for assessing an individual's sodium intake especially in relationship to health outcomes. The recommendations should be applied by scientific review committees, granting agencies, editors and journal reviewers, investigators, policymakers, and those developing and creating dietary sodium recommendations. Low-quality research on dietary sodium/salt should not be funded, conducted, or published.
Collapse
Affiliation(s)
- Norm R C Campbell
- Department of Medicine, Physiology and Pharmacology, Community Health Sciences, O'Brien Institute for Public Health and Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, Alberta, Canada
| | - Feng J He
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Monique Tan
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Francesco P Cappuccio
- Division of Health Sciences, WHO Collaborating Centre for Nutrition, Warwick Medical School, University of Warwick, Coventry, UK
| | - Bruce Neal
- The George Institute for Global Health, Sydney, New South Wales, Australia
| | - Mark Woodward
- The George Institute for Global Health, University of Oxford, Oxford, UK
| | - Mary E Cogswell
- Division for Heart Disease and Stroke Prevention, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Chamblee, Georgia
| | - Rachael McLean
- Department of Preventive and Social Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Joanne Arcand
- Faculty of Health Sciences, University of Ontario Institute of Technology, Oshawa Ontario, Canada
| | - Graham MacGregor
- Wolfson Institute of Preventive Medicine, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, London, UK
| | - Paul Whelton
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana.,Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Antti Jula
- Department of Public Health Solutions, National Institute for Health and Welfare, Turku, Finland
| | - Mary R L'Abbe
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Laura K Cobb
- Resolve to Save Lives, Vital Strategies, New York, NY
| | - Daniel T Lackland
- Department of Neurology, Division of Translational Neuroscience and Population Studies, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
50
|
Ackland GL, Brudney CS, Cecconi M, Ince C, Irwin MG, Lacey J, Pinsky MR, Grocott MPW, Mythen MG, Edwards MR, Miller TE, Miller TE, Mythen MG, Grocott MPW, Edwards MR, Ackland GL, Brudney CS, Cecconi M, Ince C, Irwin MG, Lacey J, Pinsky MR, Sanders R, Hughes F, Bader A, Thompson A, Hoeft A, Williams D, Shaw AD, Sessler DI, Aronson S, Berry C, Gan TJ, Kellum J, Plumb J, Bloomstone J, McEvoy MD, Thacker JK, Gupta R, Koepke E, Feldheiser A, Levett D, Michard F, Hamilton M. Perioperative Quality Initiative consensus statement on the physiology of arterial blood pressure control in perioperative medicine. Br J Anaesth 2019; 122:542-551. [DOI: 10.1016/j.bja.2019.01.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 01/19/2023] Open
|