Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Circ Res ; 134(10): 1234-1239, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38723029

ABSTRACT

The year 2024 marks the centennial of the initiation of the American Heart Association. Over the past 100 years, the American Heart Association has led groundbreaking discoveries in cardiovascular disease including salt sensitivity of blood pressure, which has been studied since the mid-1900s. Salt sensitivity of blood pressure is an important risk factor for cardiovascular events, but the phenotype remains unclear because of insufficient understanding of the underlying mechanisms and lack of feasible diagnostic tools. In honor of this centennial, we commemorate the initial discovery of salt sensitivity of blood pressure and chronicle the subsequent scientific discoveries and efforts to mitigate salt-induced cardiovascular disease with American Heart Association leading the way. We also highlight determinants of the pathophysiology of salt sensitivity of blood pressure in humans and recent developments in diagnostic methods and future prospects.


Subject(s)
Blood Pressure , Hypertension , Sodium Chloride, Dietary , Animals , Humans , American Heart Association/history , Blood Pressure/drug effects , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/etiology , Hypertension/etiology , Hypertension/history , Hypertension/physiopathology , Sodium Chloride, Dietary/adverse effects , Sodium Chloride, Dietary/history , United States/epidemiology , History, 20th Century , History, 21st Century
2.
Hypertension ; 81(3): 456-467, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37767696

ABSTRACT

Race is a social construct, but self-identified Black people are known to have higher prevalence and worse outcomes of hypertension than White people. This may be partly due to the disproportionate incidence of salt sensitivity of blood pressure in Black people, a cardiovascular risk factor that is independent of blood pressure and has no proven therapy. We review the multiple physiological systems involved in regulation of blood pressure, discuss what, if anything is known about the differences between Black and White people in these systems and how they affect salt sensitivity of blood pressure. The contributions of genetics, epigenetics, environment, and social determinants of health are briefly touched on, with the hope of stimulating further work in the field.


Subject(s)
Hypertension , Social Determinants of Health , Humans , Blood Pressure/genetics , Hypertension/etiology , Hypertension/genetics , Sodium Chloride, Dietary/adverse effects , Sodium Chloride , Black People/genetics
3.
Hypertension ; 81(3): 516-529, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37675576

ABSTRACT

BACKGROUND: The mechanisms by which salt increases blood pressure in people with salt sensitivity remain unclear. Our previous studies found that high sodium enters antigen-presenting cells (APCs) via the epithelial sodium channel and leads to the production of isolevuglandins and hypertension. In the current mechanistic clinical study, we hypothesized that epithelial sodium channel-dependent isolevuglandin-adduct formation in APCs is regulated by epoxyeicosatrienoic acids (EETs) and leads to salt-sensitive hypertension in humans. METHODS: Salt sensitivity was assessed in 19 hypertensive subjects using an inpatient salt loading and depletion protocol. Isolevuglandin-adduct accumulation in APCs was analyzed using flow cytometry. Gene expression in APCs was analyzed using cellular indexing of transcriptomes and epitopes by sequencing analysis of blood mononuclear cells. Plasma and urine EETs were measured using liquid chromatography-mass spectrometry. RESULTS: Baseline isolevuglandin+ APCs correlated with higher salt-sensitivity index. Isolevuglandin+ APCs significantly decreased from salt loading to depletion with an increasing salt-sensitivity index. We observed that human APCs express the epithelial sodium channel δ subunit, SGK1 (salt-sensing kinase serum/glucocorticoid kinase 1), and cytochrome P450 2S1. We found a direct correlation between baseline urinary 14,15 EET and salt-sensitivity index, whereas changes in urinary 14,15 EET negatively correlated with isolevuglandin+ monocytes from salt loading to depletion. Coincubation with 14,15 EET inhibited high-salt-induced increase in isolevuglandin+ APC. CONCLUSIONS: Isolevuglandin formation in APCs responds to acute changes in salt intake in salt-sensitive but not salt-resistant people with hypertension, and this may be regulated by renal 14,15 EET. Baseline levels of isolevuglandin+ APCs or urinary 14,15 EET may provide diagnostic tools for salt sensitivity without a protocol of salt loading.


Subject(s)
Hypertension , Lipids , Sodium Chloride, Dietary , Humans , Sodium Chloride, Dietary/metabolism , Epithelial Sodium Channels/metabolism , Sodium Chloride/metabolism , Eicosanoids , Blood Pressure/physiology
4.
Front Physiol ; 14: 1208270, 2023.
Article in English | MEDLINE | ID: mdl-37534363

ABSTRACT

Introduction: Salt sensitivity of blood pressure is a phenomenon in which blood pressure changes according to dietary sodium intake. Our previous studies found that high salt activates antigen presenting cells, resulting in the development of hypertension. The mechanisms by which salt-induced immune cell activation is regulated in salt sensitivity of blood pressure are unknown. In the current study, we investigated dietary salt-induced effects on the renin-angiotensin-aldosterone system (RAAS) gene expression in myeloid immune cells and their impact on salt sensitive hypertension in humans. Methods: We performed both bulk and single-cell sequencing analysis on immune cells with in vitro and in vivo high dietary salt treatment in humans using a rigorous salt-loading/depletion protocol to phenotype salt-sensitivity of blood pressure. We also measured plasma renin and aldosterone using radioimmunoassay. Results: We found that while in vitro high sodium exposure downregulated the expression of renin, renin binding protein and renin receptor, there were no significant changes in the genes of the renin-angiotensin system in response to dietary salt loading and depletion in vivo. Plasma renin in salt sensitive individuals tended to be lower with a blunted response to the salt loading/depletion challenge as previously reported. Discussion: These findings suggest that unlike systemic RAAS, acute changes in dietary salt intake do not regulate RAAS expression in myeloid immune cells.

5.
Hypertension ; 80(1): e1-e3, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36475860
6.
J Hypertens ; 40(10): 1859-1875, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36052518

ABSTRACT

Even though it has been more than a decade since renal denervation (RDN) was first used to treat hypertension and an intense effort on researching this therapy has been made, it is still not clear how RDN fits into the antihypertensive arsenal. There is no question that RDN lowers blood pressure (BP), it does so to an extent at best corresponding to one antihypertensive drug. The procedure has an excellent safety record. However, it remains clinically impossible to predict whose BP responds to RDN and whose does not. Long-term efficacy data on BP reduction are still unconvincing despite the recent results in the SPYRAL HTN-ON MED trial; experimental studies indicate that reinnervation is occurring after RDN. Although BP is an acceptable surrogate endpoint, there is complete lack of outcome data with RDN. Clear indications for RDN are lacking although patients with resistant hypertension, those with documented increase in activity of the sympathetic system and perhaps those who desire to take fewest medication may be considered.


Subject(s)
Antihypertensive Agents , Hypertension , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Blood Pressure , Denervation/methods , Humans , Hypertension/drug therapy , Hypertension/surgery , Kidney , Sympathectomy/methods , Treatment Outcome
7.
Circ Res ; 131(4): 328-344, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35862128

ABSTRACT

BACKGROUND: Salt sensitivity of blood pressure is an independent predictor of cardiovascular morbidity and mortality. The exact mechanism by which salt intake increases blood pressure and cardiovascular risk is unknown. We previously found that sodium entry into antigen-presenting cells (APCs) via the amiloride-sensitive epithelial sodium channel EnaC (epithelial sodium channel) leads to the formation of IsoLGs (isolevuglandins) and release of proinflammatory cytokines to activate T cells and modulate salt-sensitive hypertension. In the current study, we hypothesized that ENaC-dependent entry of sodium into APCs activates the NLRP3 (NOD [nucleotide-binding and oligomerization domain]-like receptor family pyrin domain containing 3) inflammasome via IsoLG formation leading to salt-sensitive hypertension. METHODS: We performed RNA sequencing on human monocytes treated with elevated sodium in vitro and Cellular Indexing of Transcriptomes and Epitopes by Sequencing analysis of peripheral blood mononuclear cells from participants rigorously phenotyped for salt sensitivity of blood pressure using an established inpatient protocol. To determine mechanisms, we analyzed inflammasome activation in mouse models of deoxycorticosterone acetate salt-induced hypertension as well as salt-sensitive mice with ENaC inhibition or expression, IsoLG scavenging, and adoptive transfer of wild-type dendritic cells into NLRP3 deficient mice. RESULTS: We found that high levels of salt exposure upregulates the NLRP3 inflammasome, pyroptotic and apoptotic caspases, and IL (interleukin)-1ß transcription in human monocytes. Cellular Indexing of Transcriptomes and Epitopes by Sequencing revealed that components of the NLRP3 inflammasome and activation marker IL-1ß dynamically vary with changes in salt loading/depletion. Mechanistically, we found that sodium-induced activation of the NLRP3 inflammasome is ENaC and IsoLG dependent. NLRP3 deficient mice develop a blunted hypertensive response to elevated sodium, and this is restored by the adoptive transfer of NLRP3 replete APCs. CONCLUSIONS: These findings reveal a mechanistic link between ENaC, inflammation, and salt-sensitive hypertension involving NLRP3 inflammasome activation in APCs. APC activation via the NLRP3 inflammasome can serve as a potential diagnostic biomarker for salt sensitivity of blood pressure.


Subject(s)
Hypertension , Inflammasomes , Animals , Epithelial Sodium Channels/genetics , Epitopes , Humans , Hypertension/chemically induced , Hypertension/genetics , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sodium/metabolism , Sodium Chloride/metabolism , Sodium Chloride, Dietary/adverse effects
8.
J Hum Hypertens ; 36(11): 952-959, 2022 11.
Article in English | MEDLINE | ID: mdl-35469059

ABSTRACT

The gut microbiota has recently gained attention due to its association with cardiovascular health, cancers, gastrointestinal disorders, and non-communicable diseases. One critical question is how the composition of the microbiota contributes to cardiovascular diseases (CVDs). Insightful reviews on the gut microbiota, its metabolites and the mechanisms that underlie its contribution to CVD are limited. Hence, the aim of this review was to describe linkages between the composition of the microbiota and CVD, CVD risk factors such as hypertension, diet, ageing, and sex differences. We have also highlighted potential therapies for improving the composition of the gut microbiota, which may result in better cardiovascular health.


Subject(s)
Cardiovascular Diseases , Cardiovascular System , Gastrointestinal Microbiome , Hypertension , Humans , Female , Male , Cardiovascular Diseases/etiology , Hypertension/etiology , Diet
10.
Curr Hypertens Rep ; 24(5): 145-156, 2022 05.
Article in English | MEDLINE | ID: mdl-35124768

ABSTRACT

PURPOSE OF REVIEW: The study aims to review recent advances in knowledge on the interplay between miRNAs and the sex-determining Region Y (SRY)-related high-mobility-group box 6 (Sox6) in physiology and pathophysiology, highlighting an important role in autoimmune and cardiometabolic conditions. RECENT FINDINGS: The transcription factor Sox6 is an important member of the SoxD family and plays an indispensable role in adult tissue homeostasis, regeneration, and physiology. Abnormal expression of the Sox6 gene has been implicated in several disease conditions including diabetes, cardiomyopathy, autoimmune diseases, and hypertension. Expression of Sox6 is regulated by miRNAs, which are RNAs of about 22 nucleotides, and have also been implicated in several pathophysiological conditions where Sox6 plays a role. Regulation of Sox6 by miRNAs is important in diverse physiological tissues and organs. Dysregulation of the interplay between miRNAs and Sox6 is an important determinant of various disease conditions and may be actionable for therapeutic purposes.


Subject(s)
Hypertension , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism
12.
Front Physiol ; 12: 793924, 2021.
Article in English | MEDLINE | ID: mdl-34966295

ABSTRACT

Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular morbidity and mortality that is seen in both hypertensive and normotensive populations. Insulin resistance (IR) strongly correlates with SSBP and affects nearly 50% of salt sensitive people. While the precise mechanism by which IR and SSBP relate remains elusive, several common pathways are involved in the genesis of both processes, including vascular dysfunction and immune activation. Vascular dysfunction associated with insulin resistance is characterized by loss of nitric oxide (NO)-mediated vasodilation and heightened endothelin-1 induced vasoconstriction, as well as capillary rarefaction. It manifests with increased blood pressure (BP) in salt sensitive murine models. Another common denominator in the pathogenesis of insulin resistance, hypertension, and salt sensitivity (SS) is immune activation involving pro-inflammatory cytokines like tumor necrosis factor (TNF)-α, IL-1ß, and IL-6. In the last decade, a new understanding of interstitial sodium storage in tissues such as skin and muscle has revolutionized traditional concepts of body sodium handling and pathogenesis of SS. We have shown that interstitial Na+ can trigger a T cell mediated inflammatory response through formation of isolevuglandin protein adducts in antigen presenting cells (APCs), and that this response is implicated in salt sensitive hypertension. The peroxisome proliferator-activated receptor γ (PPARγ) is a transcription factor that modulates both insulin sensitivity and BP. PPARγ agonists increase insulin sensitivity and ameliorate salt sensitivity, whereas deficiency of PPARγ results in severe insulin resistance and hypertension. These findings suggest that PPARγ plays a role in the common pathogenesis of insulin sensitivity and salt sensitivity, perhaps via effects on the immune system and vascular function. The goal of this review is to discuss those mechanisms that may play a role in both SSBP and in insulin resistance.

13.
Antioxid Redox Signal ; 35(18): 1477-1493, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34569287

ABSTRACT

Significance: Salt sensitivity of blood pressure (SSBP) is an independent risk factor for mortality and morbidity due to cardiovascular disease, and disproportionately affects blacks and women. Several mechanisms have been proposed, including exaggerated activation of sodium transporters in the kidney leading to salt retention and water. Recent Advances: Recent studies have found that in addition to the renal epithelium, myeloid immune cells can sense sodium via the epithelial Na+ channel (ENaC), which leads to activation of the nicotinamide adenine dinucleotide phosphate oxidase enzyme complex, increased fatty acid oxidation, and production of isolevuglandins (IsoLGs). IsoLGs are immunogenic and contribute to salt-induced hypertension. In addition, aldosterone-mediated activation of ENaC has been attributed to the increased SSBP in women. The goal of this review is to highlight mechanisms contributing to SSBP in blacks and women, including, but not limited to increased activation of ENaC, fatty acid oxidation, and inflammation. Critical Issues: A critical barrier to progress in management of SSBP is that its diagnosis is not feasible in the clinic and is limited to expensive and laborious research protocols, which makes it difficult to investigate. Yet without understanding the underlying mechanisms, this important risk factor remains without treatment. Future Directions: Further studies are needed to understand the mechanisms that contribute to differential blood pressure responses to dietary salt and find feasible diagnostic tools. This is extremely important and may go a long way in mitigating the racial and sex disparities in cardiovascular outcomes. Antioxid. Redox Signal. 35, 1477-1493.


Subject(s)
Hypertension , Sodium Chloride, Dietary , Black or African American , Blood Pressure , Female , Humans , Hypertension/etiology , Inflammation/complications , Oxidative Stress , Sodium
14.
Hypertension ; 78(2): 252-260, 2021 08.
Article in English | MEDLINE | ID: mdl-34232678

ABSTRACT

Salt sensitivity of blood pressure is an independent risk factor for cardiovascular mortality not only in hypertensive but also in normotensive adults. The diagnosis of salt sensitivity of blood pressure is not feasible in the clinic due to lack of a simple diagnostic test, making it difficult to investigate therapeutic strategies. Most research efforts to understand the mechanisms of salt sensitivity of blood pressure have focused on renal regulation of sodium. However, salt retention or plasma volume expansion is not different between salt-sensitive and salt-resistant individuals. In addition, over 70% of extracellular fluid is interstitial and, therefore, not directly controlled by renal salt and water excretion. We discuss in this review how the seminal work by Harry Goldblatt paved the way for our attempts at understanding the mechanisms that underlie immune activation by salt in hypertension. We describe our findings that sodium, entering antigen-presenting cells via an epithelial sodium channel, triggers a PKC (protein kinase C)- and SGK1 (serum/glucocorticoid kinase 1)-stimulated activation of nicotinamide adenine dinucleotide phosphate oxidase, which, in turn, enhances lipid oxidation with generation of highly reactive isolevuglandins. Isolevuglandins adduct to proteins, with the potential to generate degraded peptide neoantigens. Activated antigen-presenting cells increase production of the TH17 polarizing cytokines, IL (interleukin)-6, IL-1ß, and IL-23, which leads to differentiation and proliferation of IL-17A producing T cells. Our laboratory and others have shown that this cytokine contributes to hypertension. We also discuss where this sodium activation of antigen-presenting cells may occur in vivo and describe the multiple experiments, with pharmacological antagonists and knockout mice that we used to unravel this sequence of events in rodents. Finally, we describe experiments in mononuclear cells obtained from normotensive or hypertensive volunteers, which confirm that analogous processes of salt-induced immunity take place in humans.


Subject(s)
Hypertension/immunology , Kidney Diseases/immunology , Sodium Chloride, Dietary/adverse effects , Animals , Awards and Prizes , Humans , Hypertension/etiology , Kidney Diseases/etiology
16.
Circ Res ; 128(7): 908-933, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33793336

ABSTRACT

Elevated cardiovascular risk including stroke, heart failure, and heart attack is present even after normalization of blood pressure in patients with hypertension. Underlying immune cell activation is a likely culprit. Although immune cells are important for protection against invading pathogens, their chronic overactivation may lead to tissue damage and high blood pressure. Triggers that may initiate immune activation include viral infections, autoimmunity, and lifestyle factors such as excess dietary salt. These conditions activate the immune system either directly or through their impact on the gut microbiome, which ultimately produces chronic inflammation and hypertension. T cells are central to the immune responses contributing to hypertension. They are activated in part by binding specific antigens that are presented in major histocompatibility complex molecules on professional antigen-presenting cells, and they generate repertoires of rearranged T-cell receptors. Activated T cells infiltrate tissues and produce cytokines including interleukin 17A, which promote renal and vascular dysfunction and end-organ damage leading to hypertension. In this comprehensive review, we highlight environmental, genetic, and microbial associated mechanisms contributing to both innate and adaptive immune cell activation leading to hypertension. Targeting the underlying chronic immune cell activation in hypertension has the potential to mitigate the excess cardiovascular risk associated with this common and deadly disease.


Subject(s)
Hypertension/immunology , Immunity, Cellular/physiology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Antihypertensive Agents/therapeutic use , B-Lymphocytes/immunology , Complement System Proteins/immunology , Cytokines/immunology , Dendritic Cells/immunology , Drug Resistance , Female , Gastrointestinal Microbiome/immunology , Heart Disease Risk Factors , Host Microbial Interactions , Humans , Hypertension/drug therapy , Immune System Phenomena , Immunity, Innate , Inflammasomes/immunology , Inflammation/genetics , Inflammation/immunology , Macrophages/immunology , Male , Monocytes/immunology , Sex Factors , Sodium Chloride, Dietary/adverse effects , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , Virus Diseases/immunology
17.
Cardiovasc Res ; 117(5): 1358-1371, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33038226

ABSTRACT

AIMS: Prior studies have focused on the role of the kidney and vasculature in salt-induced modulation of blood pressure; however, recent data indicate that sodium accumulates in tissues and can activate immune cells. We sought to examine mechanisms by which salt causes activation of human monocytes both in vivo and in vitro. METHODS AND RESULTS: To study the effect of salt in human monocytes, monocytes were isolated from volunteers to perform several in vitro experiments. Exposure of human monocytes to elevated Na+ex vivo caused a co-ordinated response involving isolevuglandin (IsoLG)-adduct formation, acquisition of a dendritic cell (DC)-like morphology, expression of activation markers CD83 and CD16, and increased production of pro-inflammatory cytokines tumour necrosis factor-α, interleukin (IL)-6, and IL-1ß. High salt also caused a marked change in monocyte gene expression as detected by RNA sequencing and enhanced monocyte migration to the chemokine CC motif chemokine ligand 5. NADPH-oxidase inhibition attenuated monocyte activation and IsoLG-adduct formation. The increase in IsoLG-adducts correlated with risk factors including body mass index, pulse pressure. Monocytes exposed to high salt stimulated IL-17A production from autologous CD4+ and CD8+ T cells. In addition, to evaluate the effect of salt in vivo, monocytes and T cells isolated from humans were adoptively transferred to immunodeficient NSG mice. Salt feeding of humanized mice caused monocyte-dependent activation of human T cells reflected by proliferation and accumulation of T cells in the bone marrow. Moreover, we performed a cross-sectional study in 70 prehypertensive subjects. Blood was collected for flow cytometric analysis and 23Na magnetic resonance imaging was performed for tissue sodium measurements. Monocytes from humans with high skin Na+ exhibited increased IsoLG-adduct accumulation and CD83 expression. CONCLUSION: Human monocytes exhibit co-ordinated increases in parameters of activation, conversion to a DC-like phenotype and ability to activate T cells upon both in vitro and in vivo sodium exposure. The ability of monocytes to be activated by sodium is related to in vivo cardiovascular disease risk factors. We therefore propose that in addition to the kidney and vasculature, immune cells like monocytes convey salt-induced cardiovascular risk in humans.


Subject(s)
Lipid Metabolism/drug effects , Lipids , Monocytes/drug effects , NADPH Oxidases/metabolism , Sodium Chloride/pharmacology , Adoptive Transfer , Adult , Aged , Animals , Antigens, CD/metabolism , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Enzyme Activation , Female , GPI-Linked Proteins/metabolism , Humans , Immunoglobulins/metabolism , Inflammation Mediators/metabolism , Lymphocyte Activation , Male , Membrane Glycoproteins/metabolism , Mice, Transgenic , Middle Aged , Monocytes/enzymology , Monocytes/immunology , Monocytes/transplantation , Phenotype , Receptors, IgG/metabolism , Sodium Chloride, Dietary/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , CD83 Antigen
18.
Curr Hypertens Rep ; 22(10): 79, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32880753

ABSTRACT

PURPOSE OF REVIEW: Salt sensitivity of blood pressure (SSBP) is an independent predictor of death due to cardiovascular events and affects nearly 50% of the hypertensive and 25% of the normotensive population. Strong evidence indicates that reducing sodium (Na+) intake decreases blood pressure (BP) and cardiovascular events. The precise mechanisms of how dietary Na+ contributes to elevation and cardiovascular disease remain unclear. The goal of this review is to discuss mechanisms of salt-induced cardiovascular disease and how the microbiome may play a role. RECENT FINDINGS: The innate and adaptive immune systems are involved in the genesis of salt-induced hypertension. Mice fed a high-salt diet exhibit increased inflammation with a marked increase in dendritic cell (DC) production of interleukin (IL)-6 and formation of isolevuglandins (IsoLG)-protein adducts, which drive interferon-gamma (IFN-γ) and IL-17A production by T cells. While prior studies have mainly focused on the brain, kidney, and vasculature as playing a role in salt-induced hypertension, the gut is the first and largest location for Na+ absorption. Research from our group and others strongly suggests that the gut microbiome contributes to salt-induced inflammation and hypertension. Recent studies suggest that alterations in the gut microbiome contribute to salt-induced hypertension. However, the contribution of the microbiome to SSBP and its underlying mechanisms are not known. Targeting the microbiota and the associated immune cell activation could conceivably provide the much-needed therapy for SSBP.


Subject(s)
Gastrointestinal Microbiome , Hypertension , Animals , Blood Pressure , Humans , Hypertension/etiology , Inflammation , Mice , Sodium Chloride, Dietary/adverse effects
19.
Curr Hypertens Rep ; 22(10): 78, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32880756

ABSTRACT

PURPOSE OF REVIEW: With the advent of highly active antiretroviral therapy (ART), the life span of persons with HIV (PWH) has been nearly normalized. With aging, prevalence of the metabolic syndrome (MetS), including hypertension, has increased in the HIV population and exceeds that in the general population in some studies. This is due to a combination of traditional risk factors in addition to the effects attributable to the virus and ART. We review recent findings on the mechanisms contributing to MetS and hypertension in PWH, particularly those specific to the viral infection and to ART. RECENT FINDINGS: Activation of the renin-angiotensin-aldosterone system (RAAS) and chronic immune activation contribute to the development of MetS and hypertension in PWH. HIV proteins and some ART agents alter adipocyte health contributing to dyslipidemias, weight gain, and insulin resistance. HIV infection also contributes to hypertension by direct effects on the RAAS that intertwine with inflammation by the RAAS also contributing to T cell activation. Recent data suggest that in addition to current ART, therapeutic targeting of the MetS and hypertension in PWH, by interfering with the RAAS, treating insulin resistance directly or by use of immunomodulators that dampen inflammation, may be critical for preventing or treating these risk factors and to improve overall cardiovascular complications in the HIV-infected aging population.


Subject(s)
Dyslipidemias , HIV Infections , Hypertension , Insulin Resistance , Metabolic Syndrome , Aged , HIV Infections/complications , HIV Infections/drug therapy , Humans , Hypertension/complications , Hypertension/drug therapy , Metabolic Syndrome/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...