1
|
Brobak KM, Halvorsen LV, Aass HCD, Søraas CL, Aune A, Olsen E, Bergland OU, Rognstad S, Blom KB, Birkeland JAK, Høieggen A, Larstorp ACK, Solbu MD. Novel biomarkers in patients with uncontrolled hypertension with and without kidney damage. Blood Press 2024; 33:2323980. [PMID: 38606688 DOI: 10.1080/08037051.2024.2323980] [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: 11/15/2023] [Accepted: 02/20/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Estimated glomerular filtration rate (eGFR) and urine albumin/creatinine ratio (ACR) are insensitive biomarkers for early detection of hypertension-mediated organ damage (HMOD). In this nationwide cross-sectional study, we assessed potential biomarkers for early HMOD in healthy persons and patients with hypertension. We hypothesised that plasma levels of biomarkers: (1) are different between healthy controls and patients with hypertension, (2): can classify patients with hypertension according to the degree of hypertension severity. DESIGN AND METHODS Patients with hypertension prescribed ≥2 antihypertensive agents were selected from a multicentre study. Healthy controls were selected from an ongoing study of living kidney donor candidates. Uncontrolled hypertension was defined as systolic daytime ambulatory blood pressure ≥135 mmHg. Kidney HMOD was defined by ACR > 3.0 mg/mmol or eGFR < 60 mL/min/1.73 m2. Patients with hypertension were categorised into three groups: (1) controlled hypertension; (2) uncontrolled hypertension without kidney HMOD; (3) uncontrolled hypertension with kidney HMOD. Fifteen biomarkers were analysed using a Luminex bead-based immunoassay, and nine fell within the specified analytical range. RESULTS Plasma levels of Interleukin 1 receptor antagonist (IL-1RA), neutrophil gelatinase-associated lipocalin (NGAL) and uromodulin were significantly different between healthy controls (n = 39) and patients with hypertension (n = 176). In regression models, with controlled hypertension (n = 55) as the reference category, none of the biomarkers were associated with uncontrolled hypertension without (n = 59) and with (n = 62) kidney HMOD. In models adjusted for cardiovascular risk factors and eGFR, osteopontin (OPN) was associated with uncontrolled hypertension without kidney HMOD (odds ratio (OR) 1.77 (1.05-2.98), p = 0.03), and regulated upon activation normal T-cell expressed and secreted (RANTES) with uncontrolled hypertension with kidney HMOD (OR 0.57 (0.34-0.95), p = 0.03). CONCLUSIONS None of the biomarkers could differentiate our hypertension groups when established risk factors were considered. Plasma OPN may identify patients with uncontrolled hypertension at risk for kidney HMOD.
Collapse
Affiliation(s)
- Karl Marius Brobak
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Artic University of Norway, Tromsø, Norway
| | - Lene V Halvorsen
- Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Cardiovascular and Renal Research, Oslo University Hospital Ullevål, Oslo, Norway
| | | | - Camilla L Søraas
- Section for Cardiovascular and Renal Research, Oslo University Hospital Ullevål, Oslo, Norway
- Section for Environmental and Occupational Medicine, Oslo University Hospital Ullevål, Oslo, Norway
| | - Arleen Aune
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Eirik Olsen
- Clinic of Emergency Medicine and Prehospital Care, Trondheim University Hospital, Trondheim, Norway
- Department of Circulation and Medical Imaging, University of Trondheim, Trondheim, Norway
| | - Ola Undrum Bergland
- Section for Cardiovascular and Renal Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Stine Rognstad
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Cardiovascular and Renal Research, Oslo University Hospital Ullevål, Oslo, Norway
- Department of Pharmacology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Kjersti B Blom
- Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute for Experimental Medical Research, and KG Jebsen Center for Cardiac Research, Oslo University Hospital, Ullevål and University of Oslo, Oslo, Norway
| | | | - Aud Høieggen
- Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Cardiovascular and Renal Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Anne Cecilie K Larstorp
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Section for Cardiovascular and Renal Research, Oslo University Hospital Ullevål, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital Ullevål, Oslo, Norway
| | - Marit D Solbu
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Artic University of Norway, Tromsø, Norway
| |
Collapse
|
2
|
Agarwal R. Blood Pressure Control and the Prevention of Incident CKD: The Disconnect Between Cohort Studies and Randomized Trials. Am J Kidney Dis 2024:S0272-6386(24)00919-3. [PMID: 39396360 DOI: 10.1053/j.ajkd.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 10/15/2024]
Affiliation(s)
- Rajiv Agarwal
- Richard L. Roudebush VA Medical Center, Indianapolis, Indiana.
| |
Collapse
|
3
|
Kohagura K, Zamami R, Oshiro N, Shinzato Y, Uesugi N. Heterogeneous afferent arteriolopathy: a key concept for understanding blood pressure-dependent renal damage. Hypertens Res 2024:10.1038/s41440-024-01916-z. [PMID: 39379463 DOI: 10.1038/s41440-024-01916-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/02/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024]
Abstract
Hypertension, aging, and other factors are associated with arteriosclerosis and arteriolosclerosis, primary morphological features of nephrosclerosis. Although such pathological changes are not invariably linked with renal decline but are prevalent across chronic kidney disease (CKD), understanding kidney damage progression is more pragmatic than precisely diagnosing nephrosclerosis itself. Hyalinosis and medial thickening of the afferent arteriole, along with intimal thickening of small arteries, can disrupt the autoregulatory system, jeopardizing glomerular perfusion pressure given systemic blood pressure (BP) fluctuations. Consequently, such vascular lesions cause glomerular damage by inducing glomerular hypertension and ischemia at the single nephron level. Thus, the interaction between systemic BP and afferent arteriolopathy markedly influences BP-dependent renal damage progression in nephrosclerosis. Both dilated and narrowed types of afferent arteriolopathy coexist throughout the kidney, with varying proportions among patients. Therefore, optimizing antihypertensive therapy to target either glomerular hypertension or ischemia is imperative. In recent years, clinical trials have indicated that combining renin-angiotensin system inhibitors (RASis) and sodium-glucose transporter 2 inhibitors (SGLT2is) is superior to using RASis alone in slowing renal function decline, despite comparable reductions in albuminuria. The superior efficacy of SGLT2is may arise from their beneficial effects on both glomerular hypertension and renal ischemia. A comprehensive understanding of the interaction between systemic BP and heterogeneous afferent arteriolopathy is pivotal for optimizing therapy and mitigating renal decline in patients with CKD of any etiology. Therefore, in this comprehensive review, we explore the role of afferent arteriolopathy in BP-dependent renal damage.
Collapse
Affiliation(s)
- Kentaro Kohagura
- Dialysis Unit, University of the Ryukyus Hospital, Okinawa, Japan.
| | - Ryo Zamami
- Department of Cardiovascular Medicine, Nephrology and Neurology Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Nanako Oshiro
- Dialysis Unit, University of the Ryukyus Hospital, Okinawa, Japan
- Department of Cardiovascular Medicine, Nephrology and Neurology Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yuki Shinzato
- Department of Cardiovascular Medicine, Nephrology and Neurology Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Noriko Uesugi
- Department of Pathology, Fukuoka University School of Medicine, Fukuoka, Japan
| |
Collapse
|
4
|
Bautista-Pérez R, Franco M. Purinergic Receptor Antagonists: A Complementary Treatment for Hypertension. Pharmaceuticals (Basel) 2024; 17:1060. [PMID: 39204165 PMCID: PMC11357398 DOI: 10.3390/ph17081060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
The treatment of hypertension has improved in the last century; attention has been directed to restoring several altered pathophysiological mechanisms. However, regardless of the current treatments, it is difficult to control blood pressure. Uncontrolled hypertension is responsible for several cardiovascular complications, such as chronic renal failure, which is frequently observed in hypertensive patients. Therefore, new approaches that may improve the control of arterial blood pressure should be considered to prevent serious cardiovascular disorders. The contribution of purinergic receptors has been acknowledged in the pathophysiology of hypertension; this review describes the participation of these receptors in the alteration of kidney function in hypertension. Elevated interstitial ATP concentrations are essential for the activation of renal purinergic receptors; this becomes a fundamental pathway that leads to the development and maintenance of hypertension. High ATP levels modify essential mechanisms implicated in the long-term control of blood pressure, such as pressure natriuresis, the autoregulation of the glomerular filtration rate and renal blood flow, and tubuloglomerular feedback responses. Any alteration in these mechanisms decreases sodium excretion. ATP stimulates the release of vasoactive substances, causes renal function to decline, and induces tubulointerstitial damage. At the same time, a deleterious interaction involving angiotensin II and purinergic receptors leads to the deterioration of renal function.
Collapse
Affiliation(s)
- Rocio Bautista-Pérez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico;
| | - Martha Franco
- Department of Cardio-Renal Pathophysiology, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| |
Collapse
|
5
|
Brobak KM, Melsom T, Eriksen BO, Høieggen A, Norvik JV, Solbu MD. The Association between Urinary Sodium-Potassium Ratio, Kidney Function, and Blood Pressure in a Cohort from the General Population. Kidney Blood Press Res 2024; 49:184-195. [PMID: 38382490 DOI: 10.1159/000535977] [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: 06/07/2023] [Accepted: 12/07/2023] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Subclinical kidney dysfunction may contribute to salt-sensitive hypertension. We assessed the association between the urinary sodium-potassium ratio (Na/K ratio) and blood pressure (BP) in a general population cohort without diabetes, chronic kidney disease, cardiovascular disease, or treated hypertension. We investigated whether any such association was mediated by the kidney function markers measured glomerular filtration rate (mGFR), urinary albumin-creatinine ratio (ACR), and urinary epidermal growth factor-creatinine ratio (EGF-Cr). METHODS The Tromsø Study is a population-based study of inhabitants of the municipality of Tromsø, Northern Norway. Participants aged 50-62 years, without diabetes, chronic kidney disease, or cardiovascular disease, were invited to the substudy Renal Iohexol Clearance Survey in Tromsø 6 (RENIS-T6; 2007-09). For the present study, we excluded participants reporting the use of 1 or more antihypertensive agents, leaving 1,311 RENIS-T6 participants for a cross-sectional analysis. We measured office BP, 24-h ambulatory blood pressure (ABP), and mGFR using iohexol clearance. Na/K ratio, ACR, and EGF-Cr were measured in morning urine samples. RESULTS Urinary Na/K ratio was significantly associated with systolic office BP and ABP independently of cardiovascular risk factors and kidney function markers. A one-standard deviation unit increase in the Na/K ratio was associated with increased systolic ABP by 1.0 (0.3-1.6) mm Hg. Urinary Na/K ratio showed a stronger association with office BP than ABP. EGF-Cr, ACR, and mGFR did not mediate the relationship between urinary Na/K ratio and systolic BP. CONCLUSIONS In a representative sample of the middle-aged North-European population without diabetes, chronic kidney disease, cardiovascular disease, or treated hypertension, there was a consistent association between urinary Na/K ratio and BP. The association with BP was not mediated through kidney function measures, suggesting a relationship between a diet with high sodium and low potassium and higher BP regardless of kidney function.
Collapse
Affiliation(s)
- Karl Marius Brobak
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Toralf Melsom
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Bjørn Odvar Eriksen
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Aud Høieggen
- Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jon Viljar Norvik
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Marit Dahl Solbu
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
6
|
Steegh FMEG, Keijbeck AA, de Hoogt PA, Rademakers T, Houben AJHM, Reesink KD, Stehouwer CDA, Daemen MJAP, Peutz-Kootstra CJ. Capillary rarefaction: a missing link in renal and cardiovascular disease? Angiogenesis 2024; 27:23-35. [PMID: 37326760 DOI: 10.1007/s10456-023-09883-8] [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: 03/20/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023]
Abstract
Patients with chronic kidney disease (CKD) have an increased risk for cardiovascular morbidity and mortality. Capillary rarefaction may be both one of the causes as well as a consequence of CKD and cardiovascular disease. We reviewed the published literature on human biopsy studies and conclude that renal capillary rarefaction occurs independently of the cause of renal function decline. Moreover, glomerular hypertrophy may be an early sign of generalized endothelial dysfunction, while peritubular capillary loss occurs in advanced renal disease. Recent studies with non-invasive measurements show that capillary rarefaction is detected systemically (e.g., in the skin) in individuals with albuminuria, as sign of early CKD and/or generalized endothelial dysfunction. Decreased capillary density is found in omental fat, muscle and heart biopsies of patients with advanced CKD as well as in skin, fat, muscle, brain and heart biopsies of individuals with cardiovascular risk factors. No biopsy studies have yet been performed on capillary rarefaction in individuals with early CKD. At present it is unknown whether individuals with CKD and cardiovascular disease merely share the same risk factors for capillary rarefaction, or whether there is a causal relationship between rarefaction in renal and systemic capillaries. Further studies on renal and systemic capillary rarefaction, including their temporal relationship and underlying mechanisms are needed. This review stresses the importance of preserving and maintaining capillary integrity and homeostasis in the prevention and management of renal and cardiovascular disease.
Collapse
Affiliation(s)
- Floor M E G Steegh
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Anke A Keijbeck
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Patrick A de Hoogt
- Surgery, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Timo Rademakers
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Alfons J H M Houben
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Koen D Reesink
- Biomedical Engineering, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Coen D A Stehouwer
- Internal Medicine, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Mat J A P Daemen
- Department of Pathology, UMC Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Carine J Peutz-Kootstra
- Department of Pathology, Maastricht University Medical Centre+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
- Department of Pathology, Gelre Ziekenhuizen, Apeldoorn, The Netherlands.
- , Porthoslaan 39, 6213 CN, Maastricht, The Netherlands.
| |
Collapse
|
7
|
Kim GH. Primary Role of the Kidney in Pathogenesis of Hypertension. Life (Basel) 2024; 14:119. [PMID: 38255734 PMCID: PMC10817438 DOI: 10.3390/life14010119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Previous transplantation studies and the concept of 'nephron underdosing' support the idea that the kidney plays a crucial role in the development of essential hypertension. This suggests that there are genetic factors in the kidney that can either elevate or decrease blood pressure. The kidney normally maintains arterial pressure within a narrow range by employing the mechanism of pressure-natriuresis. Hypertension is induced when the pressure-natriuresis mechanism fails due to both subtle and overt kidney abnormalities. The inheritance of hypertension is believed to be polygenic, and essential hypertension may result from a combination of genetic variants that code for renal tubular sodium transporters or proteins involved in regulatory pathways. The renin-angiotensin-aldosterone system (RAAS) and sympathetic nervous system (SNS) are the major regulators of renal sodium reabsorption. Hyperactivity of either the RAAS or SNS leads to a rightward shift in the pressure-natriuresis curve. In other words, hypertension is induced when the activity of RAAS and SNS is not suppressed despite increased salt intake. Sodium overload, caused by increased intake and/or reduced renal excretion, not only leads to an expansion of plasma volume but also to an increase in systemic vascular resistance. Endothelial dysfunction is caused by an increased intracellular Na+ concentration, which inhibits endothelial nitric oxide (NO) synthase and reduces NO production. The stiffness of vascular smooth muscle cells is increased by the accumulation of intracellular Na+ and subsequent elevation of cytoplasmic Ca++ concentration. In contrast to the hemodynamic effects of osmotically active Na+, osmotically inactive Na+ stimulates immune cells and produces proinflammatory cytokines, which contribute to hypertension. When this occurs in the gut, the microbiota may become imbalanced, leading to intestinal inflammation and systemic hypertension. In conclusion, the primary cause of hypertension is sodium overload resulting from kidney dysregulation.
Collapse
Affiliation(s)
- Gheun-Ho Kim
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
| |
Collapse
|
8
|
Vogt L, Marques FZ, Fujita T, Hoorn EJ, Danser AHJ. Novel mechanisms of salt-sensitive hypertension. Kidney Int 2023; 104:690-697. [PMID: 37454911 DOI: 10.1016/j.kint.2023.06.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/08/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
A high dietary sodium-consumption level is considered the most important lifestyle factor that can be modified to help prevent an increase in blood pressure and the development of hypertension. Despite numerous studies over the past decades, the pathophysiology explaining why some people show a salt-sensitive blood pressure response and others do not is incompletely understood. Here, a brief overview of the latest mechanistic insights is provided, focusing on the mononuclear phagocytic system and inflammation, the gut-kidney axis, and epigenetics. The article also discusses the effects of 3 types of novel drugs on salt-sensitive hypertension-sodium-glucose cotransporter 2 inhibitors, nonsteroidal mineralocorticoid receptor antagonists, and aldosterone synthase inhibitors. The conclusion is that besides kidney-centered mechanisms, vasoconstrictor mechanisms are also relevant for both the understanding and treatment of this blood pressure phenotype.
Collapse
Affiliation(s)
- Liffert Vogt
- Department of Internal Medicine, Section of Nephrology, Amsterdam University Medical Centers, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, and Victorian Heart Institute, Monash University, Melbourne, Victoria, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Toshiro Fujita
- Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Tokyo, Japan
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus MC, University Medical Center, Rotterdam, Netherlands.
| |
Collapse
|
9
|
van der Weijden J, De Hoogt PA, Leufkens MME, Keijbeck AA, van Goor H, van den Heuvel MC, Cleutjens JPM, Moers C, Snoeijs MG, Navis GJ, van Londen M, Nolte IM, Berger SP, De Borst MH, Peutz-Kootstra CJ. The relationship of peritubular capillary density with glomerular volume and kidney function in living kidney donors. J Nephrol 2023; 36:2111-2124. [PMID: 37768545 PMCID: PMC10543576 DOI: 10.1007/s40620-023-01734-5] [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: 03/09/2023] [Accepted: 07/03/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Peritubular capillary rarefaction plays an important role in the progression of chronic kidney disease. Little is known about the relation between peritubular capillary density, glomerular volume and filtration rate in the healthy kidney. METHODS In this single-center study, we included 69 living kidney donors who donated between 2005 and 2008 and had representative renal biopsies available. In all donors, glomerular filtration rate was measured using 125I-Iothalamate before donation and at five years after donation. Before donation, the increase in glomerular filtration rate after dopamine stimulation was measured. Glomerular volume and peritubular capillary density were determined in biopsies taken at the time of transplantation. Pearson's correlation coefficient and linear regression were used to assess relations between parameters. RESULTS Mean donor age was 52 ± 11 years and mean measured glomerular filtration rate was 119 ± 22 mL/min before donation and 82 ± 15 mL/min at five years after donation. While peritubular capillary density (measured by either number of peritubular capillaries/50,000 μm2 or number of peritubular capillaries/tubule) was not associated with measured glomerular filtration rate before or after donation, number of peritubular capillaries/tubule was associated with the increase in measured glomerular filtration rate after dopamine stimulation (St.β = 0.33, p = 0.004), and correlated positively with glomerular volume (R = 0.24, p = 0.047). Glomerular volume was associated with unstimulated measured glomerular filtration rate before donation (St.β = 0.31, p = 0.01) and at five years (St.β = 0.30, p = 0.01) after donation, independent of age. CONCLUSIONS In summary, peritubular capillary density was not related to unstimulated kidney function before or after kidney donation, in contrast to glomerular volume. However, number of peritubular capillaries/tubule correlated with the increase in glomerular filtration rate after dopamine stimulation in healthy kidneys, and with glomerular volume. These findings suggest that peritubular capillary density and glomerular volume differentially affect kidney function in healthy living kidney donors.
Collapse
Affiliation(s)
- J van der Weijden
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands.
| | - P A De Hoogt
- Department of Vascular Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - M M E Leufkens
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - A A Keijbeck
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - H van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M C van den Heuvel
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J P M Cleutjens
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - C Moers
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M G Snoeijs
- Department of Vascular Surgery, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G J Navis
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - M van Londen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - I M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - S P Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - M H De Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box AA53, 9713 GZ, Groningen, The Netherlands
| | - C J Peutz-Kootstra
- Department of Pathology, Maastricht University Medical Center+, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
- Department of Pathology, Gelre Ziekenhuizen, Apeldoorn, The Netherlands
| |
Collapse
|
10
|
Khan MB, Scherzer R, Lewis CE, Malhotra R, Ix JH, Shlipak MG, Gutiérrez OM. Associations of Urine Biomarkers of Kidney Tubule Health With Incident Hypertension and Longitudinal Blood Pressure Change in Middle-Aged Adults: The CARDIA Study. Hypertension 2023; 80:1353-1362. [PMID: 36987923 PMCID: PMC10192098 DOI: 10.1161/hypertensionaha.123.21084] [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: 02/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Urine biomarkers of kidney tubule injury associate with incident hypertension in older adults with comorbidities, but less is known about these associations in younger adults. METHODS In 1170 participants of the CARDIA study (Coronary Artery Risk Development in Young Adults; mean age, 45 years; 40% Black people; 56% women) without hypertension, cardiovascular disease, or kidney disease at baseline, we examined associations of urine MCP-1 (monocyte chemoattractant protein-1), α1m (alpha-1-microglobulin), KIM-1 (kidney injury molecule-1), EGF (epidermal growth factor), IL (interleukin)-18, YKL-40 (chitinase-3-like protein 1), and UMOD (uromodulin) with incident hypertension (onset of systolic blood pressure [BP] ≥130 mm Hg or diastolic BP ≥80 mm Hg or initiation of hypertension medications) and longitudinal BP change in models adjusted for hypertension risk factors, estimated glomerular filtration rate, and albuminuria. RESULTS After a median 9.9 (interquartile range, 5.9-10.2) years, 376 participants developed incident hypertension. In demographic-adjusted analyses, higher tertiles of EGF associated with lower risk of incident hypertension in both Black and White participants. After multivariable adjustment, the risk of incident hypertension remained lower in tertile 2 (hazard ratio, 0.70 [95% CI, 0.50-0.97]) and tertile 3 (hazard ratio, 0.58 [0.39-0.85]) of EGF versus tertile 1. In fully adjusted models, participants in EGF tertile 3 had smaller 10-year increases in systolic (-3.4 [95% CI, -6.1 to -0.7] mm Hg) and diastolic BP (-2.6 [95% CI, -4.6 to -0.6] mm Hg) than tertile 1. Other biomarkers showed inconsistent associations with incident hypertension and BP change. CONCLUSIONS In middle-aged adults without hypertension, cardiovascular disease, or kidney disease, higher urine EGF associated with lower risk of incident hypertension and lower 10-year BP elevations.
Collapse
Affiliation(s)
- Muhammad B. Khan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Rebecca Scherzer
- Department of Kidney Health Research Collaborative, San Francisco Veterans Affairs Health Care System and University of California, San Francisco, San Francisco, CA
| | - Cora E. Lewis
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - Rakesh Malhotra
- Division of Nephrology-Hypertension, University of California, San Diego and Nephrology Section Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Joachim H. Ix
- Division of Nephrology-Hypertension, University of California, San Diego and Nephrology Section Veterans Affairs San Diego Healthcare System, San Diego, CA
| | - Michael G. Shlipak
- Department of Kidney Health Research Collaborative, San Francisco Veterans Affairs Health Care System and University of California, San Francisco, San Francisco, CA
- Department of Medicine, San Francisco Veterans Affairs Health Care System, San Francisco, CA
| | - Orlando M. Gutiérrez
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
11
|
Kurra V, Eräranta A, Paavonen T, Honkanen T, Myllymäki J, Riutta A, Tikkanen I, Lakkisto P, Mustonen J, Pörsti I. Moderate hyperuricaemia ameliorated kidney damage in a low-renin model of experimental renal insufficiency. Basic Clin Pharmacol Toxicol 2023; 132:21-32. [PMID: 36220802 PMCID: PMC10091954 DOI: 10.1111/bcpt.13806] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/06/2022] [Accepted: 10/06/2022] [Indexed: 01/06/2023]
Abstract
Uric acid has promoted renal fibrosis and inflammation in experimental studies, but some studies have shown nephroprotective effects due to alleviated oxidative stress. We studied the influence of experimental hyperuricaemia in surgically 5/6 nephrectomized rats. Three weeks after subtotal nephrectomy or sham operation, the rats were allocated to control diet or 2.0% oxonic acid (uricase inhibitor) diet for 9 weeks. Then blood, urine and tissue samples were taken, and renal morphology and oxidative stress were examined. Inflammation and fibrosis were evaluated using immunohistochemistry and real-time PCR (RT-PCR). Remnant kidney rats ingesting normal or oxonic acid diet presented with ~60% reduction of creatinine clearance and suppressed plasma renin activity. Oxonic acid diet increased plasma uric acid levels by >80 μmol/L. In remnant kidney rats, moderate hyperuricaemia decreased glomerulosclerosis, tubulointerstitial damage and kidney mast cell count, without influencing the fibrosis marker collagen I messenger RNA (mRNA) content. In both sham-operated and 5/6 nephrectomized rats, the mast cell product 11-epi-prostaglandin-F2α excretion to the urine and kidney tissue cyclooxygenase-2 (COX-2) levels were decreased. To conclude, hyperuricaemic remnant kidney rats displayed improved kidney morphology and reduced markers of oxidative stress and inflammation. Thus, moderately elevated plasma uric acid had beneficial effects on the kidney in this low-renin model of experimental renal insufficiency.
Collapse
Affiliation(s)
- Venla Kurra
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Arttu Eräranta
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Timo Paavonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Teemu Honkanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juhani Myllymäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Asko Riutta
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ilkka Tikkanen
- Minerva Institute for Medical Research, Biomedicum Helsinki 2U, Helsinki, Finland.,Abdominal Center, Nephrology, Helsinki University Hospital, Helsinki, Finland
| | - Päivi Lakkisto
- Minerva Institute for Medical Research, Biomedicum Helsinki 2U, Helsinki, Finland.,Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jukka Mustonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| | - Ilkka Pörsti
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
12
|
Brobak KM, Andreassen RM, Melsom T, Høieggen A, Norvik JV, Solbu MD. Associations of urinary orosomucoid, N-acetyl-β-D-glucosaminidase, and albumin with blood pressure and hypertension after 7 years. The Tromsø Study. Blood Press 2022; 31:270-283. [PMID: 36193001 DOI: 10.1080/08037051.2022.2128043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Purpose: Subclinical chronic kidney disease is known to exacerbate hypertension and progression of kidney damage. In order to initiate timely interventions, early biomarkers for this vicious circle are needed. Our aim was to describe the cross-sectional associations of urinary orosomucoid and urinary N-acetyl-β-D-glucosaminidase (NAG) with blood pressure and the longitudinal associations of urinary orosomucoid and NAG to hypertension after 7 years, and to compare the strength of these associations to the urinary albumin excretion (UAE).Material and methods: The Tromsø Study is a population-based, prospective study of inhabitants of the municipality of Tromsø, Northern Norway. Morning spot urine samples were collected on three consecutive days in the Tromsø 6 survey (2007-2008). We assessed the cross-sectional associations of urinary orosomucoid, NAG and UAE with blood pressure in Tromsø 6. In a cohort of participants attending Tromsø 6 and Tromsø 7 (2015-2016), we studied whether urinary biomarkers were longitudinally associated with hypertension.Results: A total of 7197 participants with a mean age of 63.5 years (SD 9.2), and a mean blood pressure of 141/78 mmHg (SD 23.0/10.6), were included in the study. Orosomucoid and UAE, but not NAG, was significantly associated with systolic and diastolic blood pressure in all the crude and multivariable cross-sectional analyses. Orosomucoid had consistently, although marginally, stronger associations with blood pressure. Incident hypertension at follow-up (Tromsø 7) was consistently significantly associated with urinary orosomucoid, but not urinary NAG or UAE. However, the standardized regression coefficients for orosomucoid were only marginally stronger than the standardized regression coefficients for ACR.Conclusion: In a cohort from the general population urine orosomucoid had a stronger cross-sectional association with blood pressure than UAE. After 7 years, urine orosomucoid showed the strongest association with incident hypertension. There were varying and weak associations between U-NAG, blood pressure and hypertension.
Collapse
Affiliation(s)
- Karl M Brobak
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway.,Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Runa M Andreassen
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway.,Helgeland Hospital Trust, Sandnessjøen, Norway
| | - Toralf Melsom
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway.,Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Aud Høieggen
- Department of Nephrology, Oslo University Hospital Ullevål, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jon V Norvik
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway.,Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Marit D Solbu
- Section of Nephrology, University Hospital of North Norway, Tromsø, Norway.,Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
13
|
Blobner BM, Kirabo A, Kashlan OB, Sheng S, Arnett DK, Becker LC, Boerwinkle E, Carlson JC, Gao Y, Gibbs RA, He J, Irvin MR, Kardia SLR, Kelly TN, Kooperberg C, McGarvey ST, Menon VK, Montasser ME, Naseri T, Redline S, Reiner AP, Reupena MS, Smith JA, Sun X, Vaidya D, Viaud-Martinez KA, Weeks DE, Yanek LR, Zhu X, Minster RL, Kleyman TR. Rare Variants in Genes Encoding Subunits of the Epithelial Na + Channel Are Associated With Blood Pressure and Kidney Function. Hypertension 2022; 79:2573-2582. [PMID: 36193739 PMCID: PMC9669116 DOI: 10.1161/hypertensionaha.121.18513] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 07/31/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The epithelial Na+ channel (ENaC) is intrinsically linked to fluid volume homeostasis and blood pressure. Specific rare mutations in SCNN1A, SCNN1B, and SCNN1G, genes encoding the α, β, and γ subunits of ENaC, respectively, are associated with extreme blood pressure phenotypes. No associations between blood pressure and SCNN1D, which encodes the δ subunit of ENaC, have been reported. A small number of sequence variants in ENaC subunits have been reported to affect functional transport in vitro or blood pressure. The effects of the vast majority of rare and low-frequency ENaC variants on blood pressure are not known. METHODS We explored the association of low frequency and rare variants in the genes encoding ENaC subunits, with systolic blood pressure, diastolic blood pressure, mean arterial pressure, and pulse pressure. Using whole-genome sequencing data from 14 studies participating in the Trans-Omics in Precision Medicine Whole-Genome Sequencing Program, and sequence kernel association tests. RESULTS We found that variants in SCNN1A and SCNN1B were associated with diastolic blood pressure and mean arterial pressure (P<0.00625). Although SCNN1D is poorly expressed in human kidney tissue, SCNN1D variants were associated with systolic blood pressure, diastolic blood pressure, mean arterial pressure, and pulse pressure (P<0.00625). ENaC variants in 2 of the 4 subunits (SCNN1B and SCNN1D) were also associated with estimated glomerular filtration rate (P<0.00625), but not with stroke. CONCLUSIONS Our results suggest that variants in extrarenal ENaCs, in addition to ENaCs expressed in kidneys, influence blood pressure and kidney function.
Collapse
Affiliation(s)
- Brandon M Blobner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ossama B Kashlan
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shaohu Sheng
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Lewis C Becker
- GeneSTAR Research Program, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jenna C Carlson
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yan Gao
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Tulane University Translational Science Institute, New Orleans, LA, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sharon LR Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Tulane University Translational Science Institute, New Orleans, LA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Stephen T McGarvey
- Department of Epidemiology and International Health Institute, Brown University School of Public Health, Providence, RI, USA
| | - Vipin K Menon
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - May E Montasser
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Take Naseri
- Department of Epidemiology and International Health Institute, Brown University School of Public Health, Providence, RI, USA
- Ministry of Health, Apia, Samoa
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Xiao Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Dhananjay Vaidya
- GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Daniel E Weeks
- Department of Biostatistics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lisa R Yanek
- GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | | | - Ryan L Minster
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Gadde S, Kalluru R, Cherukuri SP, Chikatimalla R, Dasaradhan T, Koneti J. Atrial Fibrillation in Chronic Kidney Disease: An Overview. Cureus 2022; 14:e27753. [PMID: 36106212 PMCID: PMC9445413 DOI: 10.7759/cureus.27753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2022] [Indexed: 11/12/2022] Open
Abstract
Chronic kidney disease (CKD) is a condition that can be caused due to any etiology leading to structural damage to the kidney, which can be measured by a decrease in estimated glomerular filtration rate (eGFR) and the presence of damage biomarkers for more than three months. This article has discussed the causal relationship between atrial fibrillation (AF) and CKD, a few of them being inflammation, renin-angiotensin-aldosterone system (RAAS) activation, anemia, and uremia associated with CKD. This review mentioned the clinical impact of the presence of AF in CKD patients. The presence of AF in CKD patients aggravates the renal dysfunction, which in turn adds to the generation of AF. This article explores the various pharmacological and interventional treatment modalities, including antiarrhythmics, anticoagulants, and cardiac ablation, and their complications, leading to restricted usage in CKD patients.
Collapse
|
15
|
Nepali DP, Suresh DS, Pikale DG, Jhaveri DS, Chaithanya DA, Bansal DM, Islam DR, Chanpura DA. Hypertension and the role of dietary fiber. Curr Probl Cardiol 2022; 47:101203. [DOI: 10.1016/j.cpcardiol.2022.101203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 11/03/2022]
|
16
|
Emfietzoglou M, Terentes-Printzios D, Kotronias RA, Marin F, Montalto C, De Maria GL, Banning AP. The spectrum and systemic associations of microvascular dysfunction in the heart and other organs. NATURE CARDIOVASCULAR RESEARCH 2022; 1:298-311. [PMID: 39196132 DOI: 10.1038/s44161-022-00045-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/02/2022] [Indexed: 08/29/2024]
Abstract
Microvascular dysfunction (MVD) contributes to several conditions that increase morbidity and mortality, including ischemic heart disease, heart failure, dementia, chronic kidney disease and hypertension. Consequently, MVD imposes a substantial burden on healthcare systems worldwide. In comparison to macrovascular dysfunction, MVD has been incompletely investigated, and it remains uncertain whether MVD in an organ constitutes a distinct pathology or a manifestation of a systemic disorder. Here, we summarize and appraise the techniques that are used to diagnose MVD. We review the disorders of the heart, brain and kidneys in which the role of MVD has been highlighted and summarize evidence hinting at a systemic or multi-organ nature of MVD. Finally, we discuss the benefits and limitations of implementing MVD testing in clinical practice with a focus on new interventions that are beginning to emerge.
Collapse
Affiliation(s)
| | | | | | - Federico Marin
- Oxford Heart Centre, Oxford University Hospitals, Oxford, UK
| | | | | | | |
Collapse
|
17
|
Hu J, Wang Y, Chen J, Shen Z, Song N, Zhao S, Li Y, Zou J, Zhang X, Ding X. Rationale and validation of predicting high sodium intake by spot urinary chloride in patients with chronic kidney disease. Clin Nutr ESPEN 2021; 45:284-291. [PMID: 34620330 DOI: 10.1016/j.clnesp.2021.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To analyze the rationale and evaluate the validity of spot urinary chloride or derived formulas to predict high sodium intake in patients with chronic kidney disease (CKD). METHODS We collected consecutive CKD patients at stages 1-4 who were admitted to our Nephrology department in a single center from January 01, 2014, to December 31, 2017, and tested spot and 24-hour urinary analysis on the same day. The feasibility of urinary chloride to predict urinary sodium was firstly analyzed by calculating their correlations. The validity of predicting excessive sodium intake by spot urinary sodium and chloride, two derived formulas based on spot urinary sodium or chloride, and our previous "CKDSALT" equation were accessed. We finally conducted Receiver operating characteristic (ROC) curves to compare their performance in detecting high sodium intake. RESULTS All 5204 patients were eventually analyzed. In the derivation cohort (n = 2447), a strong positive linear correlation existed between urinary sodium and chloride in both spot urine (R2 = 0.804) and 24-hour urine samples (R2 = 0.905), and two predictive equations based on spot urinary sodium or chloride were derived. In the validation cohort (n = 2757), spot urinary sodium and chloride only showed "fair" performance. However, both urinary sodium and chloride equations had a "good" performance in ICC, Pearson's correlation, Bland-Altman plots, and ROC curves, while and CKDSALT equation showed the best performance. CONCLUSIONS Spot urinary chloride is a feasible method to predict and monitor high sodium intake in CKD patients, while a novel derived formula could elevate its diagnostic accuracy.
Collapse
Affiliation(s)
- Jiachang Hu
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Yimei Wang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Jing Chen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Ziyan Shen
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Nana Song
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Shuan Zhao
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Yang Li
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Jianzhou Zou
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Xiaoyan Zhang
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Shanghai Medical Center of Kidney, Shanghai 200032, China; Shanghai Institute of Kidney and Dialysis, Shanghai 200032, China; Shanghai Key Laboratory of Kidney and Blood Purification, Shanghai 200032, China; Hemodialysis Quality Control Center of Shanghai, Shanghai 200032, China.
| |
Collapse
|
18
|
Liu L, Zhang X, Qie R, Han M, Li Q, Zhang L, Zhan S, Zhang J, Zhang C, Hong F. Dose-response association between serum uric acid levels and incident hypertension: a systematic review and meta-analysis of 17 prospective cohort studies of 32 thousand participants. Acta Cardiol 2021; 76:748-753. [PMID: 32539677 DOI: 10.1080/00015385.2020.1779476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Various magnitudes of the risk of incident hypertension (IHTN) have been reported to be associated with increased serum uric acid (SUA) levels in observational studies, however, whether a dose-response relation exists is unclear. We aimed to quantitatively evaluate the SUA-IHTN association. METHODS We searched the PubMed and Embase databases for relevant articles published prior to 21 October 2019. Random-effects models were used to estimate the summary relative risks (RRs) and 95% confidence intervals (CIs) of the IHTN risk in relation to SUA levels. We used restricted cubic splines to model the dose-response association between SUA levels and IHTN. RESULTS A total of 17 articles (17 prospective cohort studies) including 321,716 adults and 65,890 IHTN cases were identified. The pooled RR was 1.10 (95% CI 1.07-1.13; I2=90.7%; n = 17) per 1 mg/dL change in the SUA level. In addition, we found evidence of a linear and positive dose-response association between SUA levels and IHTN (Pnon-linearity = 0.069). The results of the subgroup and sensitivity analyses were consistent with those of the primary analysis. CONCLUSION These data suggested that people with higher SUA levels had a higher IHTN risk. SUA levels need to be controlled to reduce or eliminate the risk of IHTN associated with SUA levels. Clinical trial studies or diagnostic studies are needed to determine the optimal cut-off point for SUA.
Collapse
Affiliation(s)
- Leilei Liu
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Xiao Zhang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Ranran Qie
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Minghui Han
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Quanman Li
- College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Linyuan Zhang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Shaohui Zhan
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Juntao Zhang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Cailiang Zhang
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Feng Hong
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| |
Collapse
|
19
|
You S, Xu J, Wu B, Wu S, Zhang Y, Sun Y, Zhang N. Comprehensive Bioinformatics Analysis Identifies POLR2I as a Key Gene in the Pathogenesis of Hypertensive Nephropathy. Front Genet 2021; 12:698570. [PMID: 34422001 PMCID: PMC8375388 DOI: 10.3389/fgene.2021.698570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/08/2021] [Indexed: 11/13/2022] Open
Abstract
Hypertensive nephropathy (HN), mainly caused by chronic hypertension, is one of the major causes of end-stage renal disease. However, the pathogenesis of HN remains unclarified, and there is an urgent need for improved treatments. Gene expression profiles for HN and normal tissue were obtained from the Gene Expression Omnibus database. A total of 229 differentially co-expressed genes were identified by weighted gene co-expression network analysis and differential gene expression analysis. These genes were used to construct protein–protein interaction networks to search for hub genes. Following validation in an independent external dataset and in a clinical database, POLR2I, one of the hub genes, was identified as a key gene related to the pathogenesis of HN. The expression level of POLR2I is upregulated in HN, and the up-regulation of POLR2I is positively correlated with renal function in HN. Finally, we verified the protein levels of POLR2I in vivo to confirm the accuracy of our analysis. In conclusion, our study identified POLR2I as a key gene related to the pathogenesis of HN, providing new insights into the molecular mechanisms underlying HN.
Collapse
Affiliation(s)
- Shilong You
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Jiaqi Xu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Boquan Wu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Shaojun Wu
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Ying Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Naijin Zhang
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Agnoletti D, Cicero AFG, Borghi C. The Impact of Uric Acid and Hyperuricemia on Cardiovascular and Renal Systems. Cardiol Clin 2021; 39:365-376. [PMID: 34247750 DOI: 10.1016/j.ccl.2021.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The description of gout dates back almost 5000 years, and scientific interest in uric acid increased when it was found to be involved in the pathogenesis of gout. Since then, many basic and clinical studies have assessed the implications of uric acid for the oxidative system, inflammation, and cardiovascular and renal outcomes. Uric acid-lowering therapy failed to improve clinical hard outcomes in asymptomatic hyperuricemia, and it is retained in symptomatic hyperuricemia. Dietary and lifestyle modifications are critical to manage hyperuricemia. More studies are warranted to investigate the role of uric acid-lowering drugs on cardiovascular outcomes.
Collapse
Affiliation(s)
- Davide Agnoletti
- Internal Medicine Department, IRCCS Sacro Cuore Hospital, Viale Luigi Rizzardi 4, Negrar di Valpolicella (VR) 37024, Italy
| | - Arrigo F G Cicero
- Medical and Surgical Sciences Department, University of Bologna, Via Albertoni 15, Bologna 40138, Italy
| | - Claudio Borghi
- Medical and Surgical Sciences Department, University of Bologna, Via Albertoni 15, Bologna 40138, Italy.
| |
Collapse
|
21
|
Basile DP, Abais-Battad JM, Mattson DL. Contribution of Th17 cells to tissue injury in hypertension. Curr Opin Nephrol Hypertens 2021; 30:151-158. [PMID: 33394732 PMCID: PMC8221512 DOI: 10.1097/mnh.0000000000000680] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Hypertension has been demonstrated to be a chief contributor to morbidity and mortality throughout the world. Although the cause of hypertension is multifactorial, emerging evidence, obtained in experimental studies, as well as observational studies in humans, points to the role of inflammation and immunity. Many aspects of immune function have now been implicated in hypertension and end-organ injury; this review will focus upon the recently-described role of Th17 cells in this pathophysiological response. RECENT FINDINGS Studies in animal models and human genetic studies point to a role in the adaptive immune system as playing a contributory role in hypertension and renal tissue damage. Th17 cells, which produce the cytokine IL17, are strongly pro-inflammatory cells, which may contribute to tissue damage if expressed in chronic disease conditions. The activity of these cells may be enhanced by physiological factors associated with hypertension such as dietary salt or Ang II. This activity may culminate in the increased sodium retaining activity and exacerbation of inflammation and renal fibrosis via multiple cellular mechanisms. SUMMARY Th17 cells are a distinct component of the adaptive immune system that may strongly enhance pathways leading to increased sodium reabsorption, elevated vascular tone and end-organ damage. Moreover, this pathway may lend itself towards specific targeting for treatment of kidney disease and hypertension.
Collapse
Affiliation(s)
- David P Basile
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta, Georgia, USA
| |
Collapse
|
22
|
Mattson DL, Dasinger JH, Abais-Battad JM. Amplification of Salt-Sensitive Hypertension and Kidney Damage by Immune Mechanisms. Am J Hypertens 2021; 34:3-14. [PMID: 32725162 PMCID: PMC7891248 DOI: 10.1093/ajh/hpaa124] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/27/2020] [Accepted: 07/23/2020] [Indexed: 12/13/2022] Open
Abstract
Humans with salt-sensitive (SS) hypertension demonstrate increased morbidity, increased mortality, and renal end-organ damage when compared with normotensive subjects or those with salt-resistant hypertension. Increasing evidence indicates that immune mechanisms play an important role in the full development of SS hypertension and associated renal damage. Recent experimental advances and studies in animal models have permitted a greater understanding of the mechanisms of activation and action of immunity in this disease process. Evidence favors a role of both innate and adaptive immune mechanisms that are triggered by initial, immune-independent alterations in blood pressure, sympathetic activity, or tissue damage. Activation of immunity, which can be enhanced by a high-salt intake or by alterations in other components of the diet, leads to the release of cytokines, free radicals, or other factors that amplify renal damage and hypertension and mediate malignant disease.
Collapse
Affiliation(s)
- David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - John Henry Dasinger
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
23
|
Uric Acid and Hypertension: Prognostic Role and Guide for Treatment. J Clin Med 2021; 10:jcm10030448. [PMID: 33498870 PMCID: PMC7865830 DOI: 10.3390/jcm10030448] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
Abstract
The relationship between serum uric acid (SUA) and hypertension has been a subject of increasing interest since the 1870 discovery by Frederick Akbar Mahomed. Several epidemiological studies have shown a strong association between high SUA levels and the presence or the development of hypertension. Genetic analyses have found that xanthine oxidoreductase (XOR) genetic polymorphisms are associated with hypertension. However, genetic studies on urate transporters and Mendelian randomization studies failed to demonstrate a causal relationship between SUA and hypertension. Results from clinical trials on the role of urate-lowering therapy in the management of patients with hypertension are not uniform. Our study sought to analyze the prognostic and therapeutic role of SUA in the hypertensive disease, from uric acid (UA) biology to clinical trials on urate-lowering therapies.
Collapse
|
24
|
De Miguel C, Pelegrín P, Baroja-Mazo A, Cuevas S. Emerging Role of the Inflammasome and Pyroptosis in Hypertension. Int J Mol Sci 2021; 22:ijms22031064. [PMID: 33494430 PMCID: PMC7865380 DOI: 10.3390/ijms22031064] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/14/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammasomes are components of the innate immune response that have recently emerged as crucial controllers of tissue homeostasis. In particular, the nucleotide-binding domain, leucine-rich-containing (NLR) family pyrin domain containing 3 (NLRP3) inflammasome is a complex platform involved in the activation of caspase-1 and the maturation of interleukin (IL)-1β and IL-18, which are mainly released via pyroptosis. Pyroptosis is a caspase-1-dependent type of cell death that is mediated by the cleavage of gasdermin D and the subsequent formation of structurally stable pores in the cell membrane. Through these pores formed by gasdermin proteins cytosolic contents are released into the extracellular space and act as damage-associated molecular patterns, which are pro-inflammatory signals. Inflammation is a main contributor to the development of hypertension and it also is known to stimulate fibrosis and end-organ damage. Patients with essential hypertension and animal models of hypertension exhibit elevated levels of circulating IL-1β. Downregulation of the expression of key components of the NLRP3 inflammasome delays the development of hypertension and pharmacological inhibition of this inflammasome leads to reduced blood pressure in animal models and humans. Although the relationship between pyroptosis and hypertension is not well established yet, pyroptosis has been associated with renal and cardiovascular diseases, instances where high blood pressure is a critical risk factor. In this review, we summarize the recent literature addressing the role of pyroptosis and the inflammasome in the development of hypertension and discuss the potential use of approaches targeting this pathway as future anti-hypertensive strategies.
Collapse
Affiliation(s)
- Carmen De Miguel
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Correspondence: (C.D.M.); (S.C.); Tel.: +34-868-885031 (S.C.)
| | - Pablo Pelegrín
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (P.P.); (A.B.-M.)
| | - Alberto Baroja-Mazo
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (P.P.); (A.B.-M.)
| | - Santiago Cuevas
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; (P.P.); (A.B.-M.)
- Correspondence: (C.D.M.); (S.C.); Tel.: +34-868-885031 (S.C.)
| |
Collapse
|
25
|
Lee S, Choi KB, Kim SJ. The effect of uric acid and urinary sodium excretion on prehypertension: a nationwide population-based study. BMC Cardiovasc Disord 2020; 20:251. [PMID: 32460763 PMCID: PMC7251714 DOI: 10.1186/s12872-020-01535-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 05/17/2020] [Indexed: 11/10/2022] Open
Abstract
Background This study examined the effect of serum uric acid (SUA) level and urinary sodium excretion on blood pressure as well as their combined effect on prehypertension in a Korean population. Method Data from the 7th Korea National Health and Nutrition Examination Survey for adults (≥ 19 years of age) were used. The participants were classified into two groups, normotension and prehypertension, according to the JNC-7 definition. Logistic regression was carried out and adjusted for traditionally regarded confounders of blood pressure. All analyses considered a complex sampling design. A multivariate analysis was performed on subgroups defined according to their SUA level and urinary sodium excretion. Results The 4200 participants were divided into normotension (n = 2646) and prehypertension (n = 1554) groups. In the univariate analysis, patient age, male sex, concurrent comorbidity (diabetes mellitus, cardiovascular disease, stroke, dyslipidemia, and chronic kidney disease), uric acid, and urinary sodium excretion were associated with prehypertension. After adjusting for baseline covariates, both the SUA level and urinary sodium excretion were significant predictors of incident prehypertension (SUA, per 1 mg/dL increase, odds ratio [OR] 1.216, 95% confidence interval [95% CI] 1.131–1.309; urinary sodium excretion, per 1 g/day increase, OR 1.067, 95% CI 1.019–1.117). Additionally, simultaneously higher tertiles of SUA and urinary sodium excretion resulted in higher ORs for prehypertension. Conclusion Increased SUA is a significant risk marker for the development of prehypertension in normotensives. Simultaneously high SUA and urinary sodium excretion amplified the effect on the development of prehypertension. Our findings suggest that lowering SUA levels and reducing sodium intake will contribute to preventing hypertension.
Collapse
Affiliation(s)
- Shina Lee
- Department of Internal Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 158-710, Korea.
| | - Kyu Bok Choi
- Department of Internal Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 158-710, Korea
| | - Seung-Jung Kim
- Department of Internal Medicine, School of Medicine, Ewha Womans University, 1071, Anyangcheon-ro, Yangcheon-gu, Seoul, 158-710, Korea
| |
Collapse
|
26
|
Krajčoviechová A, Wohlfahrt P, Bruthans J, Šulc P, Lánská V, Borghi C, Cífková R. Longitudinal trends in the prevalence of hyperuricaemia and chronic kidney disease in hypertensive and normotensive adults. Blood Press 2020; 29:308-318. [PMID: 32425070 DOI: 10.1080/08037051.2020.1763158] [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: 10/24/2022]
Abstract
The purpose: To evaluate longitudinal trends in the prevalence of hyperuricaemia and chronic kidney disease (CKD) in Czech adults with and without arterial hypertension (HT).Materials and methods: Two independent cross-sectional surveys were performed in 2006-2009 and 2015-2018, each screening involving 1% population random sample of the general population of nine districts of the Czech Republic aged 25-64 years, stratified by age and gender. Hyperuricaemia was defined as serum uric acid ≥ 420 μmol/l in men, and ≥ 360 μmol/l in women. CKD was defined as estimated glomerular filtration rate < 60 ml/min/1.73 m2 and/or albumin/creatinine ratio ≥ 3 mg/mmol.Results: Final analyses included 3504 individuals examined in 2006-2009, and 2309 in 2015-2018. The overall prevalence of hyperuricaemia increased from 16.4% to 25.2% in men (p < 0.001), and from 7.6% to 10.9% in women (p < 0.001), whereas the overall prevalence of CKD declined from 6.8% to 3.6% in men (p = 0.001), and from 7.6% to 4.8% in women (p < 0.001). There was no interaction between HT and hyperuricaemia in either gender; the increase in hyperuricaemia prevalence was observed both in hypertensive and normotensive adults and was accompanied by the increased prevalence of abdominal obesity. Contrarily, there was an interaction between HT and CKD in both men (p < 0.001) and women (p = 0.011); the CKD prevalence declined only in hypertensive individuals, specifically in those using antihypertensive medication and was accompanied by the increased use of renin-angiotensin-aldosterone system (RAS) inhibitors and calcium channel blockers (CCBs).Conclusions: Over the period of 10 years, the overall prevalence of hyperuricaemia increased, while the prevalence of CKD decreased. An increase in the prevalence of hyperuricaemia was observed both in hypertensive and normotensive individuals and was accompanied by an increase in the prevalence of abdominal obesity. A decline in the prevalence of CKD was only observed in hypertensive individuals and was accompanied by the increased use of RAS inhibitors and CCBs.
Collapse
Affiliation(s)
- Alena Krajčoviechová
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
| | - Peter Wohlfahrt
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
| | - Jan Bruthans
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
| | - Pavel Šulc
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic
| | - Věra Lánská
- Medical Statistics Unit, Institute for Experimental and Clinical Medicine, Prague, Czech Republic
| | - Claudio Borghi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Renata Cífková
- Center for Cardiovascular Prevention, First Faculty of Medicine and Thomayer Hospital, Charles University in Prague, Prague, Czech Republic.,Department of Medicine II, Charles University in Prague, First Faculty of Medicine, Prague, Czech Republic
| |
Collapse
|
27
|
Sun T, Sakata F, Ishii T, Tawada M, Suzuki Y, Kinashi H, Katsuno T, Takei Y, Maruyama S, Mizuno M, Ito Y. Excessive salt intake increases peritoneal solute transport rate via local tonicity-responsive enhancer binding protein in subtotal nephrectomized mice. Nephrol Dial Transplant 2020; 34:2031-2042. [PMID: 30897196 DOI: 10.1093/ndt/gfz045] [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] [Received: 10/21/2018] [Accepted: 02/11/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND High peritoneal transport is associated with high mortality and technical failure in peritoneal dialysis (PD). Baseline peritoneal solute transport rate (PSTR) as measured by the peritoneal equilibration test (PET) within 6 months after PD initiation varies between patients. Sodium is reported to be stored in the skin or muscle of dialysis patients. This study investigated whether excessive salt intake in uremic mice caused peritoneal alterations without exposure to PD fluid. METHODS Sham-operated (Sham) and subtotal nephrectomized (Nx) mice were randomly given tap water or 1% sodium chloride (NaCl)-containing water for 8 weeks. PET was then performed to evaluate peritoneal function. Human mesothelial cell line Met-5A was used for in vitro studies. RESULTS We observed higher PSTR in Nx mice with 1% NaCl-containing drinking water (Nx + salt) compared with those with tap water (Nx + water), along with enhanced angiogenesis and inflammation in the peritoneum. Blockade of interleukin (IL)-6 signaling rescued peritoneal transport function in Nx + salt mice. In cultured Met-5A, additional NaCl in the medium upregulated IL-6 as well as vascular endothelial growth factor-A, associated with increased expression and nuclear translocation of tonicity-responsive enhancer binding protein (TonEBP). Knockdown of TonEBP lowered the induction caused by high tonicity. Peritoneal TonEBP expression was higher in Nx + salt mice, while removal of high-salt diet lowered TonEBP level and improved peritoneal transport function. CONCLUSIONS Excessive dietary salt intake caused peritoneal membrane functional and structural changes under uremic status. TonEBP regulated hypertonicity-related inflammatory changes and might play a crucial role in high baseline peritoneal transport.
Collapse
Affiliation(s)
- Ting Sun
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumiko Sakata
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takako Ishii
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsuhiro Tawada
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiro Suzuki
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kinashi
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Takayuki Katsuno
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| | - Yoshifumi Takei
- Department of Medicinal Biochemistry, Aichi Gakuin University School of Pharmacy, Nagoya, Japan
| | - Shoichi Maruyama
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Mizuno
- Department of Nephrology and Renal Replacement Therapy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuhiko Ito
- Department of Nephrology and Rheumatology, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
28
|
Dally M, Butler-Dawson J, Cruz A, Krisher L, Johnson RJ, Asensio C, Pilloni WD, Asturias EJ, Newman LS. Longitudinal trends in renal function among first time sugarcane harvesters in Guatemala. PLoS One 2020; 15:e0229413. [PMID: 32142520 PMCID: PMC7059928 DOI: 10.1371/journal.pone.0229413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/05/2020] [Indexed: 01/07/2023] Open
Abstract
Introduction Chronic kidney disease of unknown origin (CKDu) is an epidemic concentrated in agricultural communities in Central and South America, including young, male sugarcane harvesters. The purpose of this analysis is to understand early changes in kidney function among a cohort of first-time sugarcane harvesters and to determine risk factors for kidney function decline. Methods Joint latent class mixed models were used to model sub-population kidney function trajectory over the course of 4 years (2012–2016). Probability weighted logistic regression was used to determine personal health, community, and individual behavior risk factors associated with sub-population assignment. Data analysis occurred in 2019. Results Of 181 new workers median age 19 years old (IQR: 4), 39 (22%) were identified as having non-stable kidney function with an annual age-adjusted decline of estimated glomerular filtration rate (eGFR) of -1.0 ml/min per 1.73 m2 (95% CI: -3.4, 1.3). Kidney function (OR: 0.96; 95% CI: 0.93, 0.98), mild hypertension (OR: 5.21; 95% CI: 2.14, 13.94), and having a local home of residence (OR: 7.12; 95% CI: 2.41, 26.02) prior to employment in sugarcane were associated with non-stable eGFR sub-population assignment. Conclusions Mild hypertension may be an early indicator of the development of CKDu. A better understanding of preexisting risk factors is needed to determine why individuals are entering the workforce with reduced kidney function and elevated blood pressure and increased risk of renal function decline.
Collapse
Affiliation(s)
- Miranda Dally
- Center for Health, Work, & Environment, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| | - Jaime Butler-Dawson
- Center for Health, Work, & Environment, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Alex Cruz
- Pantaleon, Guatemala City, Guatemala
| | - Lyndsay Krisher
- Center for Health, Work, & Environment, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Richard J. Johnson
- Division of Renal Diseases and Hypertension, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | | | | | - Edwin J. Asturias
- Division of Pediatric Infectious Diseases, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, United States of America
- Center for Global Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Lee S. Newman
- Center for Health, Work, & Environment, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Environmental and Occupational Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| |
Collapse
|
29
|
Chapman CL, Schlader ZJ, Reed EL, Worley ML, Johnson BD. Renal and segmental artery hemodynamic response to acute, mild hypercapnia. Am J Physiol Regul Integr Comp Physiol 2020; 318:R822-R827. [PMID: 32130026 DOI: 10.1152/ajpregu.00035.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Profound increases (>15 mmHg) in arterial carbon dioxide (i.e., hypercapnia) reduce renal blood flow. However, a relatively brief and mild hypercapnia can occur in patients with sleep apnea or in those receiving supplemental oxygen therapy during an acute exacerbation of chronic obstructive pulmonary disease. We tested the hypothesis that a brief, mild hypercapnic exposure increases vascular resistance in the renal and segmental arteries. Blood velocity in 14 healthy adults (26 ± 4 yr; 7 women, 7 men) was measured in the renal and segmental arteries with Doppler ultrasound while subjects breathed room air (Air) and while they breathed a 3% CO2, 21% O2, 76% N2 gas mixture for 5 min (CO2). The end-tidal partial pressure of CO2 ([Formula: see text]) was measured via capnography. Mean arterial pressure (MAP) was measured beat to beat via the Penaz method. Vascular resistance in the renal and segmental arteries was calculated as MAP divided by blood velocity. [Formula: see text] increased with CO2 (Air: 45 ± 3, CO2: 48 ± 3 mmHg, P < 0.01), but there were no changes in MAP (P = 0.77). CO2 decreased blood velocity in the renal (Air: 35.2 ± 8.1, CO2: 32.2 ± 7.3 cm/s, P < 0.01) and segmental (Air: 24.2 ± 5.1, CO2: 21.8 ± 4.2 cm/s, P < 0.01) arteries and increased vascular resistance in the renal (Air: 2.7 ± 0.9, CO2: 3.0 ± 0.9 mmHg·cm-1·s, P < 0.01) and segmental (Air: 3.9 ± 1.0, CO2: 4.4 ± 1.0 mmHg·cm-1·s, P < 0.01) arteries. These data provide evidence that the kidneys are hemodynamically responsive to a mild and acute hypercapnic stimulus in healthy humans.
Collapse
Affiliation(s)
- Christopher L Chapman
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| | - Zachary J Schlader
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York.,Department of Kinesiology, School of Public Health, Indiana University, Bloomington, Indiana
| | - Emma L Reed
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| | - Morgan L Worley
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| | - Blair D Johnson
- Center for Research and Education in Special Environments, Department of Exercise and Nutrition Sciences, University at Buffalo, Buffalo, New York
| |
Collapse
|
30
|
Hu J, Wang Y, Song N, Zhang X, Teng J, Zou J, Ding X. Estimating 24-Hour Urinary Sodium Excretion From Spot Urine Samples in Chronic Kidney Disease Patients. J Ren Nutr 2020; 30:11-21. [DOI: 10.1053/j.jrn.2019.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/24/2019] [Accepted: 02/10/2019] [Indexed: 11/11/2022] Open
|
31
|
Stewart DJ, Langlois V, Noone D. Hyperuricemia and Hypertension: Links and Risks. Integr Blood Press Control 2019; 12:43-62. [PMID: 31920373 PMCID: PMC6935283 DOI: 10.2147/ibpc.s184685] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022] Open
Abstract
Hyperuricemia has long been recognized to be associated with increased cardiovascular risk, including risk of developing hypertension. Epidemiological findings suggest that the link with hypertension is stronger in children and adolescents. Uric acid acts as a strong antioxidant compound in the extracellular environment but has pro-inflammatory effects within the intracellular setting. A chronic phase of microvascular injury is known to occur after prolonged periods of hyperuricemia. This is proposed to contribute to afferent arteriolopathy and elevation of blood pressure that may become unresponsive to uric acid-lowering therapies over time. Studies have struggled to infer direct causality of hyperuricemia due to a vast number of confounders including body mass index. The aim of this review is to present the available data and highlight the need for large scale prospective randomized controlled trials in this area. At present, there is limited evidence to support a role for uric acid-lowering therapies in helping mitigate the risk of hypertension.
Collapse
Affiliation(s)
- Douglas J Stewart
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Valerie Langlois
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada.,Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| | - Damien Noone
- Division of Nephrology, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada.,Department of Paediatrics, University of Toronto, Toronto, Ontario M5G 1X8, Canada
| |
Collapse
|
32
|
Bruni C, Rosato E, Maestripieri V, Gigante A, Tesei G, Bellando-Randone S, Guiducci S, Chiostri M, El Aoufy K, Blagojevic J, Moggi-Pignone A, De Paulis A, Furst DE, Boddi M, Matucci-Cerinic M. The Renal Resistive Index in systemic sclerosis: Determinants, prognostic implication and proposal for specific age-adjusted cut-offs. Eur J Intern Med 2019; 70:43-49. [PMID: 31540806 DOI: 10.1016/j.ejim.2019.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 08/19/2019] [Accepted: 09/02/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Renal Resistive Index (RRI), reflects changes in both renal vascular and tubular-interstitial compartments and in systemic vascular compliance related to age and comorbidities. OBJECTIVES a) To investigate determinants of RRI in SSc population, b) its association with SSc-related features and c) to test its prognostic impact on organ specific worsening or death. METHODS 380 SSc patients ≥18 years were enrolled after giving informed consent. Baseline data on RRI, laboratory, instrumental and therapeutic features were retrospectively collected. Age-SSc adjusted cut-offs were created by dividing the population in age quartiles and considering RRI values >75th percentile as pathologic. Clinical follow-up was performed until last available visit or the development/worsening of specific internal organ involvement or death. RESULTS RRI was independently predicted by age and systolic pulmonary arterial pressure on Echo. Therefore, we created Age-SSc adjusted pathologic RRI cut-offs, which were significantly associated with various disease related skin and lung fibrotic manifestations, as well as vasculopathic complications. After a mean follow-up of 3.6 ± 2.6 years, RRI was one of the independent predictors (together with modified Rodnan skin score, interstitial lung disease, presence of dyspnoea and late nailfold-videocapillaroscopy pattern) for mortality, with 0.68 as best cut-off (sensitivity 88.5%, specificity 50.9%). CONCLUSION If corroborated, Renal Resistive Index cut-offs might be used to evaluate renal and extrarenal involvement in SSc and could serve as predictors of mortality.
Collapse
Affiliation(s)
- Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy; Department Cardio-Thorax-Vascular Medicine, Division of General Cardiology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy.
| | - Edoardo Rosato
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Vanessa Maestripieri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Antonietta Gigante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Giulia Tesei
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy; Department of Geriatric Medicine, Division of Rheumatology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy; Department of Geriatric Medicine, Division of Rheumatology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Marco Chiostri
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Khadija El Aoufy
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy
| | - Jelena Blagojevic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy; Department of Geriatric Medicine, Division of Rheumatology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Alberto Moggi-Pignone
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; Department of Internal Medicine, Division of Internal Medicine Unit III, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| | - Amato De Paulis
- Department of Translational Medical Sciences, Center for Basic and Clinical Immunology Research (CISI), WAO Center of Excellence, University Federico II, Naples, Italy
| | - Daniel E Furst
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy; Department of Geriatric Medicine, Division of Rheumatology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy; Department of Medicine, Division of Rheumatology, University of California at Los Angeles, USA; University of Washington, Seattle, WA, USA
| | - Maria Boddi
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; University of Washington, Seattle, WA, USA
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Via Delle Oblate 4, 50134 Florence, Italy; Department of Geriatric Medicine, Division of Rheumatology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
| |
Collapse
|
33
|
Liddell TS, Bassett R, Link DK. Hypertension management in patients with chronic kidney disease. Nurse Pract 2019; 44:34-40. [PMID: 31688350 DOI: 10.1097/01.npr.0000605512.81315.63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Managing hypertension, especially when accompanied by chronic kidney disease, is challenging. These different but related conditions are complicated by differing guidelines. NPs can safely prescribe antihypertensive treatments, which reduce hypertension and the risk of associated comorbidities, such as kidney failure, stroke, myocardial infarction, and vascular disease.
Collapse
Affiliation(s)
- Toddra S Liddell
- Toddra S. Liddell is an NP at Vanderbilt University Medical Center, Division of Nephrology and Hypertension, Nashville, Tenn. Robin Bassett is an NP at Advent Health, Daytona Beach, Fla., and Davita Dialysis, Ormond Beach, Fla. Denise K. Link is a PA at The University of Texas Southwestern Medical Center, Department of Nephrology, Dallas, Tex
| | | | | |
Collapse
|
34
|
Abstract
Sodium intake is undoubtedly indispensable for normal body functions but can be detrimental when taken in excess of dietary requirements. The consequences of excessive salt intake are becoming increasingly clear as high salt consumption persists across the globe. Salt has long been suspected to promote the development of hypertension and cardiovascular diseases and is now also recognized as a potential modulator of inflammatory and autoimmune diseases through its direct and indirect effects on immune cells. The finding that, in addition to the kidneys, other organs such as the skin regulate sodium levels in the body prompted new hypotheses, including the concept that skin-resident macrophages might participate in tissue sodium regulation through their interactions with lymphatic vessels. Moreover, immune cells such as macrophages and different T cell subsets are found in sodium-rich interstitial microenvironments, where sodium levels modulate their function. Alterations to the intestinal bacterial community induced by excess dietary salt represent another relevant axis whereby salt indirectly modulates immune cell function. Depending on the inflammatory context, sodium might either contribute to protective immunity (for example, by enhancing host responses against cutaneous pathogens) or it might contribute to immune dysregulation and promote the development of cardiovascular and autoimmune diseases.
Collapse
|
35
|
Franco M, Pérez-Méndez O, Kulthinee S, Navar LG. Integration of purinergic and angiotensin II receptor function in renal vascular responses and renal injury in angiotensin II-dependent hypertension. Purinergic Signal 2019; 15:277-285. [PMID: 31183668 PMCID: PMC6635571 DOI: 10.1007/s11302-019-09662-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/30/2019] [Indexed: 12/24/2022] Open
Abstract
Glomerular arteriolar vasoconstriction and tubulointerstitial injury are observed before glomerular damage occurs in models of hypertension. High interstitial ATP concentrations, caused by the increase in arterial pressure, alter renal mechanisms involved in the long-term control of blood pressure, autoregulation of glomerular filtration rate and blood flow, tubuloglomerular feedback (TGF) responses, and sodium excretion. Elevated ATP concentrations and augmented expression of P2X receptors have been demonstrated under a genetic background or induction of hypertension with vasoconstrictor peptides. In addition to the alterations of the microcirculation in the hypertensive kidney, the vascular actions of elevated intrarenal angiotensin II levels may be mitigated by the administration of broad purinergic P2 antagonists or specific P2Y12, P2X1, and P2X7 receptor antagonists. Furthermore, the prevention of tubulointerstitial infiltration with immunosuppressor compounds reduces the development of salt-sensitive hypertension, indicating that tubulointerstitial inflammation is essential for the development and maintenance of hypertension. Inflammatory cells also express abundant purinergic receptors, and their activation by ATP induces cytokine and growth factor release that in turn contributes to augment tubulointerstitial inflammation. Collectively, the evidence suggests a pathophysiological activation of purinergic P2 receptors in angiotensin-dependent hypertension. Coexistent increases in intrarenal angiotensin II and activates Ang II AT1 receptors, which interacts with over-activated purinergic receptors in a complex manner, suggesting convergence of their post-receptor signaling processes.
Collapse
Affiliation(s)
- Martha Franco
- Department of Nephrology, Renal Pathophysiology Laboratory, Instituto Nacional de Cardiología “Ignacio Chávez”, Juan Badiano No.1, 14080 Mexico City, DF Mexico
| | - Oscar Pérez-Méndez
- Department Molecular Biology, Instituto Nacional de Cardiología “Ignacio Chávez”, Mexico City, Mexico
| | - Supaporn Kulthinee
- Department of Physiology and Hypertension and Renal Center, Tulane University School of Medicine, New Orleans, LA USA
- Department of Cardiovascular and Thoracic Technology, Chulabhorn International College of Medicine, Thammasat University, Rangsit, Pathum Thani Thailand
| | - L. Gabriel Navar
- Department of Physiology and Hypertension and Renal Center, Tulane University School of Medicine, New Orleans, LA USA
| |
Collapse
|
36
|
|
37
|
Abstract
Genetic variants in APOL1, encoding apolipoprotein L1, are major drivers of glomerular disease in peoples of sub-Saharan African descent. APOL1-associated primary glomerular diseases include focal segmental glomerulosclerosis, human immunodeficiency virus-associated nephropathies, and arterionephrosclerosis. Other conditions where APOL1 variants affect outcomes include membranous nephropathy, lupus nephritis, diabetic nephropathy, preeclampsia, and kidney transplant. In focal segmental glomerulosclerosis, APOL1 variants are associated with upregulation of RNA encoding chemokine C-X-C motif receptor 3 ligands and ubiquitin D; the significance of these findings remains unclear but may provide valuable insight into disease mechanisms.
Collapse
|
38
|
Abstract
INTRODUCTION Hypertension is caused by increased cardiac output and/or increased peripheral resistance. Areas covered: The various mechanisms affecting cardiac output/peripheral resistance involved in the development of essential hypertension are covered. These include genetics; sympathetic nervous system overactivity; renal mechanisms: excess sodium intake and pressure natriuresis; vascular mechanisms: endothelial cell dysfunction and the nitric oxide pathway; hormonal mechanisms: the renin-angiotensin-aldosterone system (RAAS); obesity, obstructive sleep apnea (OSA); insulin resistance and metabolic syndrome; uric acid; vitamin D; gender differences; racial, ethnic, and environmental factors; increased left ventricular ejection force and hypertension and its association with increased basal sympathetic activity - cortical connections. Expert commentary: Maximum association of hypertension is found with sympathetic overactivity which is directly or indirectly involved in different mechanisms of hypertension including RAAS, OSA, obesity, etc.. It is not overt sympathetic activity but disturbed basal sympathetic tone. Basal sympathetic tone arises from hypothalamus; possibly affected by cortical influences. Therefore, hypertension is not merely a disease of circulatory system alone. Its pathogenesis involves alteration in ANS (autonomic nervous system) and likely in cortical-hypothalamic connections. Assessment of ANS and cortical-hypothalamic connections may be required for better understanding of hypertension.
Collapse
Affiliation(s)
- Tarun Saxena
- a Department of Internal Medicine , Mittal Hospital and Research Centre , Ajmer , India
| | - Azeema Ozefa Ali
- a Department of Internal Medicine , Mittal Hospital and Research Centre , Ajmer , India
| | - Manjari Saxena
- b Department Yoga and Physical education , Mittal Hospital and Research Centre , Ajmer , India
| |
Collapse
|
39
|
Vio CP, Salas D, Cespedes C, Diaz-Elizondo J, Mendez N, Alcayaga J, Iturriaga R. Imbalance in Renal Vasoactive Enzymes Induced by Mild Hypoxia: Angiotensin-Converting Enzyme Increases While Neutral Endopeptidase Decreases. Front Physiol 2018; 9:1791. [PMID: 30618804 PMCID: PMC6297360 DOI: 10.3389/fphys.2018.01791] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022] Open
Abstract
Chronic hypoxia has been postulated as one of the mechanisms involved in salt-sensitive hypertension and chronic kidney disease (CKD). Kidneys have a critical role in the regulation of arterial blood pressure through vasoactive systems, such as the renin-angiotensin and the kallikrein-kinin systems, with the angiotensin-converting enzyme (ACE) and kallikrein being two of the main enzymes that produce angiotensin II and bradykinin, respectively. Neutral endopeptidase 24.11 or neprilysin is another enzyme that among its functions degrade vasoactive peptides including angiotensin II and bradykinin, and generate angiotensin 1-7. On the other hand, the kidneys are vulnerable to hypoxic injury due to the active electrolyte transportation that requires a high oxygen consumption; however, the oxygen supply is limited in the medullary regions for anatomical reasons. With the hypothesis that the chronic reduction of oxygen under normobaric conditions would impact renal vasoactive enzyme components and, therefore; alter the normal balance of the vasoactive systems, we exposed male Sprague-Dawley rats to normobaric hypoxia (10% O2) for 2 weeks. We then processed renal tissue to identify the expression and distribution of kallikrein, ACE and neutral endopeptidase 24.11 as well as markers of kidney damage. We found that chronic hypoxia produced focal damage in the kidney, mainly in the cortico-medullary region, and increased the expression of osteopontin. Moreover, we observed an increase of ACE protein in the brush border of proximal tubules at the outer medullary region, with increased mRNA levels. Kallikrein abundance did not change significantly with hypoxia, but a tendency toward reduction was observed at protein and mRNA levels. Neutral endopeptidase 24.11 was localized in proximal tubules, and was abundantly expressed under normoxic conditions, which markedly decreased both at protein and mRNA levels with chronic hypoxia. Taken together, our results suggest that chronic hypoxia produces focal kidney damage along with an imbalance of key components of the renal vasoactive system, which could be the initial steps for a long-term contribution to salt-sensitive hypertension and CKD.
Collapse
Affiliation(s)
- Carlos P Vio
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Daniela Salas
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Cespedes
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jessica Diaz-Elizondo
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia Mendez
- Department of Physiology, Center for Aging and Regeneration CARE UC, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institute of Anatomy, Histology, and Pathology, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Julio Alcayaga
- Laboratorio de Fisiología Celular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Department of Physiology, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
40
|
Cao W, Shi M, Wu L, Yang Z, Yang X, Liu H, Xu X, Liu Y, Wilcox CS, Hou FF. A renal-cerebral-peripheral sympathetic reflex mediates insulin resistance in chronic kidney disease. EBioMedicine 2018; 37:281-293. [PMID: 30429087 PMCID: PMC6286258 DOI: 10.1016/j.ebiom.2018.10.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/18/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023] Open
Abstract
Background Insulin resistance (IR) complicates chronic kidney disease (CKD). We tested the hypothesis that CKD activates a broad reflex response from the kidneys and the white adipose tissue to impair peripheral glucose uptake and investigated the role of salt intake in this process. Methods 5/6-nephrectomized rats were administered normal- or high-salt for 3 weeks. Conclusions were tested in 100 non-diabetic patients with stage 3–5 CKD. Findings High-salt in 5/6-nephrectomized rats decreased insulin-stimulated 2-deoxyglucose uptake >25% via a sympathetic nervous system (SNS) reflex that linked the IR to reactive oxygen species (ROS) and the renin-angiotensin system (RAS) in brain and peripheral tissues. Salt-loading in CKD enhanced inflammation in adipose tissue and skeletal muscle, and enhanced the impairment of insulin signaling and Glut4 trafficking. Denervation of the kidneys or adipose tissue or deafferentation of adipose tissue improved IR >40%. In patients with non-diabetic CKD, IR was positively correlated with salt intake after controlling for cofounders (r = 0.334, P = 0.001) and was linked to activation of the RAS/SNS and to impaired glucose uptake in adipose tissue and skeletal muscle, all of which depended on salt intake. Interpretation CKD engages a renal/adipose-cerebral-peripheral sympathetic reflex that activates the RAS/ROS axes to promote IR via local inflammation and impaired Glut4 trafficking that are enhanced by high-salt intake. The findings point to a role for blockade of RAS or α-and-β-adrenergic receptors to reduce IR in patients with CKD. Fund National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Wei Cao
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Meng Shi
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Liling Wu
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Zhichen Yang
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Xiaobing Yang
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Hongfa Liu
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Xin Xu
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Youhua Liu
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China
| | - Christopher S Wilcox
- Division of Nephrology and Hypertension, Georgetown University Medical Central, 3800 Reservoir Road, NW, 6 PHC Bldg, F6003, Washington, DC 20007, USA.
| | - Fan Fan Hou
- Division of Nephrology, Nanfang Hospital, 1838 North Guangzhou Avenue, Guangzhou 510515, PR China..
| |
Collapse
|
41
|
Ajayi S, Adebiyi A, Kadiri S. Increased urinary sodium excretion is associated with systolic blood pressure in first degree relatives of hypertensive patients in Ibadan, Southwestern Nigeria. Pan Afr Med J 2018; 31:168. [PMID: 31086621 PMCID: PMC6488237 DOI: 10.11604/pamj.2018.31.168.16611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/24/2018] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Understanding the risk factors of hypertension has led to a better understanding of the pathogenesis, prevention and reduction in morbidity of hypertension. It is well known that offsprings of hypertensive parents have an increased risk of developing hypertension. It is therefore necessary to explore the physiological differences between normotensive patients with and without a positive family history of hypertension with respect to their urinary excretion of sodium. METHODS This study was carried out at the University College Hospital, Ibadan Nigeria, to determine if normotensive patients with a positive family history of hypertension are different with respect to their urinary excretion of electrolytes and blood pressure. It examined the relationship between 24-hour urinary excretion of sodium, chloride and potassium, urea and creatinine and blood pressure in subjects with and without family history of hypertension. It was a case-control study of sixty-two subjects: normotensive patients' first degree relatives of primary hypertensive patients and normotensive patients without positive family history. RESULTS The mean (SD) systolic blood pressures for subjects with and without family history of hypertension were significantly different: 120.0(22.25) and 105.0(17.50) respectively, (p=0.001). The mean arterial blood pressures were significantly different: 86.4(10.2) mmHg and 80.1(8.1) mmHg respectively (p=0.010). The mean (SD) 24-hour urinary excretion of sodium for normotensive patients with and without positive family history of hypertension were 180.5 (45.50) mEq/L, and 156.0(36.25) mEq/L respectively. Systolic blood pressure and 24-hour urinary excretion of sodium was also higher in normotensive subjects with a positive family history of hypertension. CONCLUSION Systolic blood pressure and twenty-four hour urinary excretion of sodium were higher in normotensive subjects with a positive family history of hypertension than in those without a family history of hypertension.
Collapse
Affiliation(s)
- Samuel Ajayi
- Nephrology Unit, Department of Medicine, University College Hospital and College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adewole Adebiyi
- Cardiology Unit, Department of Medicine, University College Hospital and College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Solomon Kadiri
- Nephrology Unit, Department of Medicine, University College Hospital and College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
42
|
Kurtz TW, DiCarlo SE, Pravenec M, Morris RC. Changing views on the common physiologic abnormality that mediates salt sensitivity and initiation of salt-induced hypertension: Japanese research underpinning the vasodysfunction theory of salt sensitivity. Hypertens Res 2018; 42:6-18. [DOI: 10.1038/s41440-018-0122-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 07/26/2018] [Accepted: 07/27/2018] [Indexed: 12/24/2022]
|
43
|
Haas ME, Aragam KG, Emdin CA, Bick AG, Hemani G, Davey Smith G, Kathiresan S. Genetic Association of Albuminuria with Cardiometabolic Disease and Blood Pressure. Am J Hum Genet 2018; 103:461-473. [PMID: 30220432 PMCID: PMC6174360 DOI: 10.1016/j.ajhg.2018.08.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/03/2018] [Indexed: 02/07/2023] Open
Abstract
Excretion of albumin in urine, or albuminuria, is associated with the development of multiple cardiovascular and metabolic diseases. However, whether pathways leading to albuminuria are causal for cardiometabolic diseases is unclear. We addressed this question using a Mendelian randomization framework in the UK Biobank, a large population-based cohort. We first performed a genome-wide association study for albuminuria in 382,500 individuals and identified 32 new albuminuria loci. We constructed albuminuria genetic risk scores and tested for association with cardiometabolic diseases. Genetically elevated albuminuria was strongly associated with increased risk of hypertension (1.38 OR; 95% CI, 1.27-1.50 per 1 SD predicted increase in albuminuria, p = 7.01 × 10-14). We then examined bidirectional associations of albuminuria with blood pressure which suggested that genetically elevated albuminuria led to higher blood pressure (2.16 mmHg systolic blood pressure; 95% CI, 1.51-2.82 per 1 SD predicted increase in albuminuria, p = 1.22 × 10-10) and that genetically elevated blood pressure led to more albuminuria (0.005 SD; 95% CI 0.004-0.006 per 1 mmHg predicted increase in systolic blood pressure, p = 2.45 × 10-13). These results support the existence of a feed-forward loop between albuminuria and blood pressure and imply that albuminuria could increase risk of cardiovascular disease through blood pressure. Moreover, they suggest therapies that target albuminuria-increasing processes could have antihypertensive effects that are amplified through inhibition of this feed-forward loop.
Collapse
Affiliation(s)
- Mary E Haas
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Krishna G Aragam
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Connor A Emdin
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Alexander G Bick
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Gibran Hemani
- Medical Research Council Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Sekar Kathiresan
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
44
|
Aziz F, Clark D, Garg N, Mandelbrot D, Djamali A. Hypertension guidelines: How do they apply to kidney transplant recipients. Transplant Rev (Orlando) 2018; 32:225-233. [DOI: 10.1016/j.trre.2018.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/05/2018] [Accepted: 06/17/2018] [Indexed: 12/28/2022]
|
45
|
Stehouwer CDA. Microvascular Dysfunction and Hyperglycemia: A Vicious Cycle With Widespread Consequences. Diabetes 2018; 67:1729-1741. [PMID: 30135134 DOI: 10.2337/dbi17-0044] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/18/2018] [Indexed: 11/13/2022]
Abstract
Microvascular and metabolic physiology are tightly linked. This Perspective reviews evidence that 1) the relationship between hyperglycemia and microvascular dysfunction (MVD) is bidirectional and constitutes a vicious cycle; 2) MVD in diabetes affects many, if not all, organs, which may play a role in diabetes-associated comorbidities such as depression and cognitive impairment; and 3) MVD precedes, and contributes to, hyperglycemia in type 2 diabetes (T2D) through impairment of insulin-mediated glucose disposal and, possibly, insulin secretion. Obesity and adverse early-life exposures are important drivers of MVD. MVD can be improved through weight loss (in obesity) and through exercise. Pharmacological interventions to improve MVD are an active area of investigation.
Collapse
Affiliation(s)
- Coen D A Stehouwer
- Department of Internal Medicine and CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre+, Maastricht, the Netherlands
| |
Collapse
|
46
|
Abstract
Tubular injury sensitizes glomeruli to injury. We review potential mechanisms of this tubuloglomerular cross talk. In the same nephron, tubular injury can cause stenosis of the glomerulotubular junction and finally result in atubular glomeruli. Tubular injury also affects glomerular filtration function through tubuloglomerular feedback. Progenitor cells, that is, parietal epithelial cells and renin positive cells, can be involved in repair of injured glomeruli and also may be modulated by tubular injury. Loss of nephrons induces additional workload and stress on remaining nephrons. Hypoxia and activation of the renin-angiotensin-aldosterone system induced by tubular injury also modulate tubuloglomerular cross talk. Therefore, effective therapies in chronic kidney disease may need to aim to interrupt this deleterious tubuloglomerular cross talk.
Collapse
Affiliation(s)
- Jiayi Wang
- 1 Division of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China.,2 Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jianyong Zhong
- 2 Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,3 Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hai-Chun Yang
- 2 Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,3 Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Agnes B Fogo
- 2 Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,3 Division of Pediatric Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
47
|
Andrade L, Rodrigues CE, Gomes SA, Noronha IL. Acute Kidney Injury as a Condition of Renal Senescence. Cell Transplant 2018; 27:739-753. [PMID: 29701108 PMCID: PMC6047270 DOI: 10.1177/0963689717743512] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Acute kidney injury (AKI), characterized by a sharp drop in glomerular filtration, continues to be a significant health burden because it is associated with high initial mortality, morbidity, and substantial health-care costs. There is a strong connection between AKI and mechanisms of senescence activation. After ischemic or nephrotoxic insults, a wide range of pathophysiological events occur. Renal tubular cell injury is characterized by cell membrane damage, cytoskeleton disruption, and DNA degradation, leading to tubular cell death by necrosis and apoptosis. The senescence mechanism involves interstitial fibrosis, tubular atrophy, and capillary rarefaction, all of which impede the morphological and functional recovery of the kidneys, suggesting a strong link between AKI and the progression of chronic kidney disease. During abnormal kidney repair, tubular epithelial cells can assume a senescence-like phenotype. Cellular senescence can occur as a result of cell cycle arrest due to increased expression of cyclin kinase inhibitors (mainly p21), downregulation of Klotho expression, and telomere shortening. In AKI, cellular senescence is aggravated by other factors including oxidative stress and autophagy. Given this scenario, the main question is whether AKI can be repaired and how to avoid the senescence process. Stem cells might constitute a new therapeutic approach. Mesenchymal stem cells (MSCs) can ameliorate kidney injury through angiogenesis, immunomodulation, and fibrosis pathway blockade, as well as through antiapoptotic and promitotic processes. Young umbilical cord–derived MSCs are better at increasing Klotho levels, and thus protecting tissues from senescence, than are adipose-derived MSCs. Umbilical cord–derived MSCs improve glomerular filtration and tubular function to a greater degree than do those obtained from adult tissue. Although senescence-related proteins and microRNA are upregulated in AKI, they can be downregulated by treatment with umbilical cord–derived MSCs. In summary, stem cells derived from young tissues, such as umbilical cord–derived MSCs, could slow the post-AKI senescence process.
Collapse
Affiliation(s)
- Lucia Andrade
- 1 Laboratory of Basic Science LIM-12, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Camila E Rodrigues
- 1 Laboratory of Basic Science LIM-12, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Samirah A Gomes
- 2 Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| | - Irene L Noronha
- 2 Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo, School of Medicine, São Paulo, Brazil
| |
Collapse
|
48
|
Yamamoto R, Imai E, Maruyama S, Yokoyama H, Sugiyama H, Nitta K, Tsukamoto T, Uchida S, Takeda A, Sato T, Wada T, Hayashi H, Akai Y, Fukunaga M, Tsuruya K, Masutani K, Konta T, Shoji T, Hiramatsu T, Goto S, Tamai H, Nishio S, Shirasaki A, Nagai K, Yamagata K, Hasegawa H, Yasuda H, Ichida S, Naruse T, Fukami K, Nishino T, Sobajima H, Tanaka S, Akahori T, Ito T, Yoshio T, Katafuchi R, Fujimoto S, Okada H, Ishimura E, Kazama JJ, Hiromura K, Mimura T, Suzuki S, Saka Y, Sofue T, Suzuki Y, Shibagaki Y, Kitagawa K, Morozumi K, Fujita Y, Mizutani M, Shigematsu T, Kashihara N, Sato H, Matsuo S, Narita I, Isaka Y. Regional variations in immunosuppressive therapy in patients with primary nephrotic syndrome: the Japan nephrotic syndrome cohort study. Clin Exp Nephrol 2018; 22:1266-1280. [PMID: 29679356 DOI: 10.1007/s10157-018-1579-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/25/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND The lack of high-quality clinical evidences hindered broad consensus on optimal therapies for primary nephrotic syndromes. The aim of the present study was to compare prevalence of immunosuppressive drug use in patients with primary nephrotic syndrome across 6 regions in Japan. METHODS Between 2009 and 2010, 380 patients with primary nephrotic syndrome in 56 hospitals were enrolled in a prospective cohort study [Japan Nephrotic Syndrome Cohort Study (JNSCS)], including 141, 151, and 38 adult patients with minimal change disease (MCD), membranous nephropathy (MN), and focal segmental glomerulosclerosis (FSGS), respectively. Their clinical characteristics were compared with those of patients registered in a large nationwide registry of kidney biopsies [Japan Renal Biopsy Registry (J-RBR)]. The regional prevalence of use of each immunosuppressive drug was assessed among adult MCD, MN, and FSGS patients who underwent immunosuppressive therapy in the JNSCS (n = 139, 127, and 34, respectively). Predictors of its use were identified using multivariable-adjusted logistic regression models. RESULTS The clinical characteristics of JNSCS patients were comparable to those of J-RBR patients, suggesting that the JNSCS included the representatives in the J-RBR. The secondary major immunosuppressive drugs were intravenous methylprednisolone [n = 33 (24.6%), 24 (19.7%), and 9 (28.1%) in MCD, MN, and FSGS, respectively] and cyclosporine [n = 25 (18.7%), 62 (50.8%), and 16 (50.0%), respectively]. The region was identified as a significant predictor of use of intravenous methylprednisolone in MCD and MN patients. CONCLUSION Use of intravenous methylprednisolone for MCD and MN differed geographically in Japan. Its efficacy should be further evaluated in a well-designed trial.
Collapse
Affiliation(s)
- Ryohei Yamamoto
- Health and Counseling Center, Osaka University, 1-17 Machikaneyama-cho, Toyonaka, Osaka, 560-0043, Japan
| | - Enyu Imai
- Nakayamadera Imai Clinic, 2-8-18 Nakayamadera, Takarazuka, Hyogo, 665-0861, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hitoshi Yokoyama
- Department of Nephrology, Kanazawa Medical Univeristy School of Medicine, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa, 920-0293, Japan
| | - Hitoshi Sugiyama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikatacho, Kita-ku, Okayama, Okayama, 700-8558, Japan
| | - Kosaku Nitta
- Department of Medicine, Kidney Center, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Tatsuo Tsukamoto
- Division of Nephrology and Dialysis, Kitano Hospital, Tazuke Kofukai Medical Research Institute, 2-4-20 Ogimachi, Kita-ku, Osaka, Osaka, 530-8480, Japan
| | - Shunya Uchida
- Department of Internal Medicine, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo, 173-8606, Japan
| | - Asami Takeda
- Kidney Disease Center, Japanese Red Cross Nagoya Daini Hospital, 2-9 Myokencho, Showa-ku, Nagoya, Aichi, 466-8650, Japan
| | - Toshinobu Sato
- Department of Nephrology, JCHO Sendai Hospital, 3-16-1 Tsutsumi-machi, Aoba-ku, Sendai, Miyagi, 981-8501, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Hiroki Hayashi
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukakecho, Toyoake, Aichi, 470-1192, Japan
| | - Yasuhiro Akai
- First Department of Internal Medicine, Nara Medical University, 840 Shijocho, Kashihara, Nara, 634-8522, Japan
| | - Megumu Fukunaga
- Division of Nephrology, Department of Internal Medicine, Toyonaka Municipal Hospital, 4-14-1 Shibaharacho, Toyonaka, Osaka, 560-8565, Japan
| | - Kazuhiko Tsuruya
- Department of Integrated Therapy for Chronic Kidney Disease, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka, 812-8582, Japan
| | - Kosuke Masutani
- Division of Nephrology and Rheumatology, Department of Internal Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, Fukuoka, 814-0180, Japan
| | - Tsuneo Konta
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University School of Medicine, 2-2 Iida-Nishi, Yamagata-shi, Yamagata, Yamagata, 990-9585, Japan
| | - Tatsuya Shoji
- Department of Kidney Disease and Hypertension, Osaka General Medical Center, 3-1-56 Bandaihigashi, Sumiyoshi-ku, Osaka, Osaka, 558-8558, Japan
| | - Takeyuki Hiramatsu
- Department of Nephrology, Konan Kosei Hospital, 137 Omatsubara, Takayacho, Konan, Aichi, 483-8704, Japan
| | - Shunsuke Goto
- Division of Nephrology and Kidney Center, Kobe University Graduate School of Medicine, 7-5-1 Kusunokicho, Cuho-ku, Kobe, Hyogo, 650-0017, Japan
| | - Hirofumi Tamai
- Department of Nephrology, Anjo Kosei hospital, 28 Higashihirokute, Anjocho, Anjo, Aichi, 446-8602, Japan
| | - Saori Nishio
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Arimasa Shirasaki
- Department of Nephrology, Ichinomiya Municipal Hospital, 2-2-22 Bunkyo, Ichinomiya, Aichi, 491-8558, Japan
| | - Kojiro Nagai
- Department of Nephrology, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15, Kuramoto-cho, Tokushima, 770-8503, Japan
| | - Kunihiro Yamagata
- Department of Nephrology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Hajime Hasegawa
- Department of Nephrology and Hypertension, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe, Saitama, 350-850, Japan
| | - Hidemo Yasuda
- Internal Medicine 1, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Shizunori Ichida
- Department of Nephrology, Japanese Red Cross Nagoya Daiichi Hospitail, 3-35 Michishitacho, Nakamura-ku, Nagoya, Aichi, 453-8511, Japan
| | - Tomohiko Naruse
- Department of Nephrology, Kasugai Municipal Hospital, 1-1-1 Takakicho, Kasugai, Aichi, 486-8510, Japan
| | - Kei Fukami
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, 67 Asahimachi, Kurume, Fukuoka, 830-0011, Japan
| | - Tomoya Nishino
- Department of Nephrology, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Nagasaki, 852-8501, Japan
| | - Hiroshi Sobajima
- Department of Diabetology and Nephrology, Ogaki Municipal Hospital, 4-86 Minaminokawacho, Ogaki, Gifu, 503-8502, Japan
| | - Satoshi Tanaka
- Department of Nephrology, Shizuoka General Hospital, 4-27-1 Kitaando, Aoi-ku, Shizuoka, Shizuoak, 420-8527, Japan
| | - Toshiyuki Akahori
- Department of Nephrology, Chutoen General Medical Center, 1-1 Shobugaike, Kakegawa, Shizuoka, 436-8555, Japan
| | - Takafumi Ito
- Division of Nephrology, Shimane University Hospital, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Terada Yoshio
- Department of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kochi University, Okocho Kohasu, Nankoku, Kochi, 783-8505, Japan
| | - Ritsuko Katafuchi
- Kideny Unit, National Fukuoka Higashi Medical Center, 1-1-1 Chidori, Koga, Fukuoka, 811-3195, Japan
| | - Shouichi Fujimoto
- Department of Hemovascular Medicine and Artificial Organs, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotakecho, Miyazaki, Miyazaki, 889-1692, Japan
| | - Hirokazu Okada
- Department of Nephrology, Saitama Medical University, 38 Morohongo, Moroyama, Iruma, Saitama, 350-0495, Japan
| | - Eiji Ishimura
- Department of Nephrology, Osaka City University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka, 545-8585, Japan
| | - Junichiro James Kazama
- Department of Nephrology and Hypertension, Fukushima Medical University School of Medicine, 1 Hikariga-oka, Fukushima-city, Fukushima, 960-1295, Japan
| | - Keiju Hiromura
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, 3-39-22 Showa-matchi, Maebashi, Gunma, 371-8511, Japan
| | - Tetsushi Mimura
- Department of Nephrology, Gifu Prefectural Tajimi Hospital, 5-161 Maebatacho, Tajimi, Gifu, 507-8522, Japan
| | - Satashi Suzuki
- Department of Nephrology, Kainan Hospital, 396 Minamihonden, Maegasucho, Yatomi, Aichi, 498-8502, Japan
| | - Yosuke Saka
- Department of Nephrology, Yokkaichi Municipal Hospital, Yokkaichi, 2-2-37 Shibata, Yokkaichi, Mie, 510-8567, Japan
| | - Tadashi Sofue
- Department of Cardiorenal and Cerebrovascular Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Yusuke Suzuki
- Division of Nephrology, Department of Internal Medicine, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Yugo Shibagaki
- Division of Nephrology and Hypertension, Department of Internal Medicine, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-000, Japan
| | - Kiyoki Kitagawa
- Division of Internal Medicine, National Hospital Organization Kanazawa Medical Center, 1-1 Shimoishibikimachi, Kanazawa, Ishikawa, 920-8650, Japan
| | - Kunio Morozumi
- Department of Nephrology, Masuko Memorial Hospital, 35-28 Takegashicho, Nakamura-ku, Nagoya, Aichi, 453-0016, Japan
| | - Yoshiro Fujita
- Department of Nephrology, Chubu Rosai Hospital, 1-10-6 Komei, Minato-ku, Nagoya, Aichi, 455-8530, Japan
| | - Makoto Mizutani
- Department of Nephrology, Handa City Hospital, 2-29 Toyocho, Handa, Aichi, 475-8599, Japan
| | - Takashi Shigematsu
- Department of Nephrology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-8509, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, 577 Matsushima, Kurashiki, Osakayama, 701-0192, Japan
| | - Hiroshi Sato
- Department of Nephrology, Endocrinology, and Vascular Medicine, Tohoku Univeristy Gradaute School of Medicine, 1-1 Seiryo-cho, Aoba-ku, Sendai, Miyagi, 980-8574, Japan
| | - Seiichi Matsuo
- Department of Nephrology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, 757 Ichibancho, Asahimachi-dori, Chuo Ward, Niigata, Niigata, 951-8510, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, 2-2-D11 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
49
|
Lopez Gelston CA, Balasubbramanian D, Abouelkheir GR, Lopez AH, Hudson KR, Johnson ER, Muthuchamy M, Mitchell BM, Rutkowski JM. Enhancing Renal Lymphatic Expansion Prevents Hypertension in Mice. Circ Res 2018; 122:1094-1101. [PMID: 29475981 DOI: 10.1161/circresaha.118.312765] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/20/2018] [Accepted: 02/21/2018] [Indexed: 01/12/2023]
Abstract
RATIONALE Hypertension is associated with renal infiltration of activated immune cells; however, the role of renal lymphatics and immune cell exfiltration is unknown. OBJECTIVE We tested the hypotheses that increased renal lymphatic density is associated with 2 different forms of hypertension in mice and that further augmenting renal lymphatic vessel expansion prevents hypertension by reducing renal immune cell accumulation. METHODS AND RESULTS Mice with salt-sensitive hypertension or nitric oxide synthase inhibition-induced hypertension exhibited significant increases in renal lymphatic vessel density and immune cell infiltration associated with inflammation. Genetic induction of enhanced lymphangiogenesis only in the kidney, however, reduced renal immune cell accumulation and prevented hypertension. CONCLUSIONS These data demonstrate that renal lymphatics play a key role in immune cell trafficking in the kidney and blood pressure regulation in hypertension.
Collapse
Affiliation(s)
| | | | | | - Alexandra H Lopez
- From the Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Kayla R Hudson
- From the Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Eric R Johnson
- From the Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Mariappan Muthuchamy
- From the Department of Medical Physiology, Texas A&M College of Medicine, College Station
| | - Brett M Mitchell
- From the Department of Medical Physiology, Texas A&M College of Medicine, College Station.
| | - Joseph M Rutkowski
- From the Department of Medical Physiology, Texas A&M College of Medicine, College Station
| |
Collapse
|
50
|
Fernández-Llama P, Calero F. [How does salt intake influence blood pressure? Associated aetiopathogenic mechanisms]. HIPERTENSION Y RIESGO VASCULAR 2017; 35:S1889-1837(17)30105-8. [PMID: 29254634 DOI: 10.1016/j.hipert.2017.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 10/18/2022]
Abstract
Abundant evidence from epidemiological and experimental studies has established a link between salt and blood pressure. However, there is heterogeneity in the blood pressure responses of humans to changes in sodium intake. Those individuals in whom a severe, abrupt change in salt intake causes the least change in arterial pressure and are termed salt-resistant, whereas in those in whom this leads to large changes in blood pressure, are called salt sensitive. Classically, Guyton's theory of the pressure-natriuresis phenomenon has been accepted to explain the pressor effect of salt, as well as the fundamental role played by the different protein sodium transporters of the renal tubules. In recent years, new theories have emerged pointing to the possible role of the immune system and the existence of a third sodium store in the body as aetiopathogenic factors.
Collapse
Affiliation(s)
- P Fernández-Llama
- Unidad de Hipertensión, Servicio de Nefrología, Fundació Puigvert, Universitat Autònoma de Barcelona (UAB), Institut d'Investigació Biomèdica Sant Pau, Barcelona, España.
| | - F Calero
- Unidad de Hipertensión, Servicio de Nefrología, Fundació Puigvert, Universitat Autònoma de Barcelona (UAB), Institut d'Investigació Biomèdica Sant Pau, Barcelona, España
| |
Collapse
|