1
|
Connolly BJ, Saxton SN. Recent updates on the influence of iron and magnesium on vascular, renal, and adipose inflammation and possible consequences for hypertension. J Hypertens 2024; 42:1848-1861. [PMID: 39258532 PMCID: PMC11451934 DOI: 10.1097/hjh.0000000000003829] [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: 02/06/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
The inflammatory status of the kidneys, vasculature, and perivascular adipose tissue (PVAT) has a significant influence on blood pressure and hypertension. Numerous micronutrients play an influential role in hypertension-driving inflammatory processes, and recent reports have provided bases for potential targeted modulation of these micronutrients to reduce hypertension. Iron overload in adipose tissue macrophages and adipocytes engenders an inflammatory environment and may contribute to impaired anticontractile signalling, and thus a treatment such as chelation therapy may hold a key to reducing blood pressure. Similarly, magnesium intake has proven to greatly influence inflammatory signalling and concurrent hypertension in both healthy animals and in a model for chronic kidney disease, demonstrating its potential clinical utility. These findings highlight the importance of further research to determine the efficacy of micronutrient-targeted treatments for the amelioration of hypertension and their potential translation into clinical application.
Collapse
Affiliation(s)
- Benjamin J Connolly
- Divison of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | | |
Collapse
|
2
|
Xu B, Dissanayake LV, Levchenko V, Zietara A, Kravtsova O, Staruschenko A. Deletion of Kcnj16 altered transcriptomic and metabolomic profiles of Dahl salt-sensitive rats. iScience 2024; 27:110901. [PMID: 39328933 PMCID: PMC11424968 DOI: 10.1016/j.isci.2024.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/06/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The inwardly rectifying K+ channel Kir5.1 (Kcnj16) is essential in renal salt handling and blood pressure control. However, the underlying mechanisms are not fully understood. Here, we integrated transcriptomics and metabolomics to comprehensively profile the changes in genes and metabolites in the Dahl salt-sensitive (SS) rat lacking Kcnj16 to identify potential mechanisms. Consistent with the phenotype of knockout (KO) rats, the transcriptomic profile predicted reduced blood pressure, kidney damage, and increased ion transport. Canonical pathway analysis suggested activation of metabolic-related pathways while suppression of immune response-related pathways in KO rats. Untargeted metabolomic analysis revealed different metabolic profiles between wild-type (WT) and KO rats. Integration of transcriptomic and metabolomic profiles suggested altered tricarboxylic acid (TCA) cycle, amino acid metabolism, and reactive oxygen species (ROS) metabolism that are related to SS hypertension. In conclusion, besides increased ion transport, our data suggest suppressed immune response-related and altered metabolic-related pathways of SS rats lacking Kir5.1.
Collapse
Affiliation(s)
- Biyang Xu
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Adrian Zietara
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Olha Kravtsova
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, USA
- James A. Haley Veteran's Hospital, Tampa, FL, USA
| |
Collapse
|
3
|
Zhou G, Gan L, Zhao B, Fang F, Liu H, Chen X, Huang J. Adding salt to foods and risk of psoriasis: A prospective cohort study. J Autoimmun 2024; 147:103259. [PMID: 38823158 DOI: 10.1016/j.jaut.2024.103259] [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: 01/31/2024] [Revised: 04/26/2024] [Accepted: 05/16/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND High salt intake may play a critical role in the etiology of psoriasis. Yet, evidence on the association of high salt intake with risk of psoriasis is limited. OBJECTIVE To estimate the association between frequency of adding salt to foods and risk of psoriasis. METHODS We conducted a prospective cohort study of 433,788 participants from the UK Biobank. Hazard ratios (HRs) and their 95 % confidence intervals (CIs) for risk of psoriasis in relation to frequency of adding salt to foods were estimated using multivariable Cox proportional hazards models. We further evaluated the joint association of adding salt to foods and genetic susceptibility with risk of psoriasis. We conducted a mediation analysis to assess how much of the effect of adding salt to foods on risk of psoriasis was mediated through several selected mediators. RESULTS During a median of 14.0 years of follow-up, 4279 incident cases of psoriasis were identified. In the multivariable-adjusted model, a higher frequency of adding salt to foods was significantly associated with an increased risk of psoriasis ("always" versus "never/rarely" adding salt to foods, HR = 1.25, 95 % CI: 1.10, 1.41). The observed positive association was generally similar across subgroups. In the joint association analysis, we observed that participants with a high genetic risk (above the second tertile) and the highest frequency of adding salt to foods experienced 149 % higher risk of psoriasis, when compared with participants with a low genetic risk (below the first tertile) and the lowest frequency of adding salt to foods (HR = 2.49, 95 % CI: 2.05, 3.02). Mediation analysis revealed that 1.8 %-3.2 % of the positive association between frequency of adding salt and risk of psoriasis was statistically significantly mediated by obesity and inflammatory biomarkers such as C-reactive protein and systemic immune-inflammation index (all P values < 0.004). CONCLUSIONS Our study demonstrated a positive association between frequency of adding salt to foods and risk of psoriasis. The positive association was independent of multiple other risk factors, and may be partially mediated through obesity and inflammation.
Collapse
Affiliation(s)
- Guowei Zhou
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Laboratory of Medical Big Data Application Technology (Central South University), Changsha, China; Furong Laboratory, Changsha, China
| | - Lu Gan
- Furong Laboratory, Changsha, China; National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Public Health, Central South University, Changsha, China; CSU-Sinocare Research Center for Nutrition and Metabolic Health, Changsha, China
| | - Bin Zhao
- Furong Laboratory, Changsha, China; National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Public Health, Central South University, Changsha, China; CSU-Sinocare Research Center for Nutrition and Metabolic Health, Changsha, China
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Laboratory of Medical Big Data Application Technology (Central South University), Changsha, China; Furong Laboratory, Changsha, China.
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Clinical Research Center for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Engineering Laboratory of Medical Big Data Application Technology (Central South University), Changsha, China; Furong Laboratory, Changsha, China.
| | - Jiaqi Huang
- Furong Laboratory, Changsha, China; National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China; Xiangya School of Public Health, Central South University, Changsha, China; CSU-Sinocare Research Center for Nutrition and Metabolic Health, Changsha, China.
| |
Collapse
|
4
|
Welling PA, Little R, Al-Qusairi L, Delpire E, Ellison DH, Fenton RA, Grimm PR. Potassium-Switch Signaling Pathway Dictates Acute Blood Pressure Response to Dietary Potassium. Hypertension 2024; 81:1044-1054. [PMID: 38465625 PMCID: PMC11023808 DOI: 10.1161/hypertensionaha.123.22546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Potassium (K+)-deficient diets, typical of modern processed foods, increase blood pressure (BP) and NaCl sensitivity. A K+-dependent signaling pathway in the kidney distal convoluted tubule, coined the K+ switch, that couples extracellular K+ sensing to activation of the thiazide-sensitive NaCl cotransporter (NCC) and NaCl retention has been implicated, but causality has not been established. METHODS To test the hypothesis that small, physiological changes in plasma K+ (PK+) are translated to BP through the switch pathway, a genetic approach was used to activate the downstream switch kinase, SPAK (SPS1-related proline/alanine-rich kinase), within the distal convoluted tubule. The CA-SPAK (constitutively active SPS1-related proline/alanine-rich kinase mice) were compared with control mice over a 4-day PK+ titration (3.8-5.1 mmol) induced by changes in dietary K+. Arterial BP was monitored using radiotelemetry, and renal function measurements, NCC abundance, phosphorylation, and activity were made. RESULTS As PK+ decreased in control mice, BP progressively increased and became sensitive to dietary NaCl and hydrochlorothiazide, coincident with increased NCC phosphorylation and urinary sodium retention. By contrast, BP in CA-SPAK mice was elevated, resistant to the PK+ titration, and sensitive to hydrochlorothiazide and salt at all PK+ levels, concomitant with sustained and elevated urinary sodium retention and NCC phosphorylation and activity. Thus, genetically locking the switch on drives NaCl sensitivity and prevents the response of BP to potassium. CONCLUSIONS Low K+, common in modern ultraprocessed diets, presses the K+-switch pathway to turn on NCC activity, increasing sodium retention, BP, and salt sensitivity.
Collapse
Affiliation(s)
- Paul A. Welling
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Robert Little
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - Lama Al-Qusairi
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, USA
| | - David H. Ellison
- Department of Medicine, Division of Nephrology, Oregon Health Science Center, Portland, Oregon, US
| | - Robert A. Fenton
- Department of Biomedicine, University of Aarhus, Aarhus, Denmark
| | - P. Richard Grimm
- Department of Medicine, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, USA
| |
Collapse
|
5
|
AlShanableh Z, Ray EC. Magnesium in hypertension: mechanisms and clinical implications. Front Physiol 2024; 15:1363975. [PMID: 38665599 PMCID: PMC11044701 DOI: 10.3389/fphys.2024.1363975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Hypertension is associated with increased risk of cardiovascular disease and death. Evidence suggests that Mg2+ depletion contributes to hypertension. It is estimated that 25% or more of the United States population experiences chronic, latent Mg2+ depletion. This review explores mechanisms by which Mg2+ influences blood pressure, modifying risk of hypertension and complicating its treatment. Mechanisms addressed include effects upon i) sympathetic tone, via the modulation of N-methyl-D-aspartate (NMDA) receptor and N-type Ca2+ channel activity, influencing catecholamine release from sympathetic nerve endings; ii) vascular tone, via alteration of L-type Ca2+ and endothelial nitric oxide synthase (eNOS) activity and prostacyclin release; iii) renal K+ handling, influencing systemic K+ balance and potentially indirectly influencing blood pressure; iv) aldosterone secretion from the adrenal cortex; and v) modulation of pro-hypertensive inflammatory processes in dendritic cells and macrophages, including activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome and stimulation of isolevuglandin (IsoLG) production. Discovery of these mechanisms has furthered our understanding of the pathogenesis of hypertension, with implications for treatment and has highlighted the role of Mg2+ balance in hypertension and cardiovascular disease.
Collapse
Affiliation(s)
| | - Evan C. Ray
- Renal-Electrolyte Division, UPMC and University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
6
|
Abstract
Excessive salt intake raises blood pressure, but the implications of this observation for human health have remained contentious. It has also been recognized for many years that potassium intake may mitigate the effects of salt intake on blood pressure and possibly on outcomes such as stroke. Recent large randomized intervention trials have provided strong support for the benefits of replacing salt (NaCl) with salt substitute (75% NaCl, 25% KCl) on hard outcomes, including stroke. During the same period of time, major advances have been made in understanding how the body senses and tastes salt, and how these sensations drive intake. Additionally, new insights into the complex interactions between systems that control sodium and potassium excretion by the kidneys, and the brain have highlighted the existence of a potassium switch in the kidney distal nephron. This switch seems to contribute importantly to the blood pressure-lowering effects of potassium intake. In recognition of these evolving data, the United States Food and Drug Administration is moving to permit potassium-containing salt substitutes in food manufacturing. Given that previous attempts to reduce salt consumption have not been successful, this new approach has a chance of improving health and ending the 'Salt Wars'.
Collapse
Affiliation(s)
- Robert Little
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- LeDucq Transatlantic Network of Excellence
| | - David H. Ellison
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, Oregon, USA
- Oregon Clinical & Translational Research Institute, Oregon Health & Science University, Portland, Oregon, USA
- LeDucq Transatlantic Network of Excellence
- VA Portland Health Care System, Portland, OR
| |
Collapse
|
7
|
Bagordo D, Rossi GP, Delles C, Wiig H, Rossitto G. Tangram of Sodium and Fluid Balance. Hypertension 2024; 81:490-500. [PMID: 38084591 PMCID: PMC10863667 DOI: 10.1161/hypertensionaha.123.19569] [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] [Indexed: 01/05/2024]
Abstract
Homeostasis of fluid and electrolytes is a tightly controlled physiological process. Failure of this process is a hallmark of hypertension, chronic kidney disease, heart failure, and other acute and chronic diseases. While the kidney remains the major player in the control of whole-body fluid and electrolyte homeostasis, recent discoveries point toward more peripheral mechanisms leading to sodium storage in tissues, such as skin and muscle, and a link between this sodium and a range of diseases, including the conditions above. In this review, we describe multiple facets of sodium and fluid balance from traditional concepts to novel discoveries. We examine the differences between acute disruption of sodium balance and the longer term adaptation in chronic disease, highlighting areas that cannot be explained by a kidney-centric model alone. The theoretical and methodological challenges of more recently proposed models are discussed. We acknowledge the different roles of extracellular and intracellular spaces and propose an integrated model that maintains fluid and electrolyte homeostasis and can be distilled into a few elemental players: the microvasculature, the interstitium, and tissue cells. Understanding their interplay will guide a more precise treatment of conditions characterized by sodium excess, for which primary aldosteronism is presented as a prototype.
Collapse
Affiliation(s)
- Domenico Bagordo
- Emergency and Hypertension Unit, Dipartimento di Medicina (DIMED), Università degli Studi di Padova, Italy (D.B., G.P.R., G.R.)
| | - Gian Paolo Rossi
- Emergency and Hypertension Unit, Dipartimento di Medicina (DIMED), Università degli Studi di Padova, Italy (D.B., G.P.R., G.R.)
| | - Christian Delles
- School of Cardiovascular & Metabolic Health, University of Glasgow, United Kingdom (G.R., C.D.)
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Norway (H.W.)
| | - Giacomo Rossitto
- Emergency and Hypertension Unit, Dipartimento di Medicina (DIMED), Università degli Studi di Padova, Italy (D.B., G.P.R., G.R.)
- School of Cardiovascular & Metabolic Health, University of Glasgow, United Kingdom (G.R., C.D.)
| |
Collapse
|
8
|
Sriperumbuduri S, Welling P, Ruzicka M, Hundemer GL, Hiremath S. Potassium and Hypertension: A State-of-the-Art Review. Am J Hypertens 2024; 37:91-100. [PMID: 37772757 DOI: 10.1093/ajh/hpad094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023] Open
Abstract
Hypertension is the single most important and modifiable risk factor for cardiovascular morbidity and mortality worldwide. Non pharmacologic interventions, in particular dietary modifications have been established to decrease blood pressure (BP) and hypertension related adverse cardiovascular events. Among those dietary modifications, sodium intake restriction dominates guidelines from professional organizations and has garnered the greatest attention from the mainstream media. Despite guidelines and media exhortations, dietary sodium intake globally has not noticeably changed over recent decades. Meanwhile, increasing dietary potassium intake has remained on the sidelines, despite similar BP-lowering effects. New research reveals a potential mechanism of action, with the elucidation of its effect on natriuresis via the potassium switch effect. Additionally, potassium-substituted salt has been shown to not only reduce BP, but also reduce the risk for stroke and cardiovascular mortality. With these data, we argue that the focus on dietary modification should shift from a sodium-focused to a sodium- and potassium-focused approach with an emphasis on intervention strategies which can easily be implemented into clinical practice.
Collapse
Affiliation(s)
- Sriram Sriperumbuduri
- Division of Nephrology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Paul Welling
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Marcel Ruzicka
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Canada
| | - Gregory L Hundemer
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Canada
| | - Swapnil Hiremath
- Division of Nephrology, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, Canada
| |
Collapse
|
9
|
Jeong S, Hunter SD, Cook MD, Grosicki GJ, Robinson AT. Salty Subjects: Unpacking Racial Differences in Salt-Sensitive Hypertension. Curr Hypertens Rep 2024; 26:43-58. [PMID: 37878224 PMCID: PMC11414742 DOI: 10.1007/s11906-023-01275-z] [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] [Accepted: 10/10/2023] [Indexed: 10/26/2023]
Abstract
PURPOSE OF REVIEW To review underlying mechanisms and environmental factors that may influence racial disparities in the development of salt-sensitive blood pressure. RECENT FINDINGS Our group and others have observed racial differences in diet and hydration, which may influence salt sensitivity. Dietary salt elicits negative alterations to the gut microbiota and immune system, which may increase hypertension risk, but little is known regarding potential racial differences in these physiological responses. Antioxidant supplementation and exercise offset vascular dysfunction following dietary salt, including in Black adults. Furthermore, recent work proposes the role of racial differences in exposure to social determinants of health, and differences in health behaviors that may influence risk of salt sensitivity. Physiological and environmental factors contribute to the mechanisms that manifest in racial differences in salt-sensitive blood pressure. Using this information, additional work is needed to develop strategies that can attenuate racial disparities in salt-sensitive blood pressure.
Collapse
Affiliation(s)
- Soolim Jeong
- Neurovascular Physiology Laboratory (NVPL), School of Kinesiology, Auburn University, Auburn, AL, 36849, USA
| | - Stacy D Hunter
- Department of Health & Human Performance, Texas State University, San Marcos, TX, 78666, USA
| | - Marc D Cook
- Department of Kinesiology, North Carolina Agriculture and Technology State University, Greensboro, NC, 27411, USA
| | - Gregory J Grosicki
- Biodynamics and Human Performance Center, Georgia Southern University (Armstrong Campus), Savannah, GA, 31419, USA
| | - Austin T Robinson
- Neurovascular Physiology Laboratory (NVPL), School of Kinesiology, Auburn University, Auburn, AL, 36849, USA.
| |
Collapse
|
10
|
Pitzer Mutchler A, Huynh L, Patel R, Lam T, Bain D, Jamison S, Kirabo A, Ray EC. The role of dietary magnesium deficiency in inflammatory hypertension. Front Physiol 2023; 14:1167904. [PMID: 37293263 PMCID: PMC10244581 DOI: 10.3389/fphys.2023.1167904] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 06/10/2023] Open
Abstract
Nearly 30% of adults consume less than the estimated average daily requirement of magnesium (Mg2+), and commonly used medications, such as diuretics, promote Mg2+ deficiency. Higher serum Mg2+ levels, increased dietary Mg2+ in-take, and Mg2+ supplementation are each associated with lower blood pressure, suggesting that Mg2+-deficiency contributes to the pathogenesis of hypertension. Antigen-presenting cells, such as monocytes and dendritic cells, are well-known to be involved in the pathogenesis of hypertension. In these cells, processes implicated as necessary for increased blood pressure include activation of the NLRP3 inflammasome, IL-1β production, and oxidative modification of fatty acids such as arachidonic acid, forming isolevuglandins (IsoLGs). We hypothesized that increased blood pressure in response to dietary Mg2+-depletion leads to increased NLRP3, IL-1β, and IsoLG production in antigen presenting cells. We found that a Mg2+-depleted diet (0.01% Mg2+ diet) increased blood pressure in mice compared to mice fed a 0.08% Mg2+ diet. Mg2+-depleted mice did not exhibit an increase in total body fluid, as measured by quantitative magnetic resonance. Plasma IL-1β concentrations were increased (0.13 ± 0.02 pg/mL vs. 0.04 ± 0.02 pg/mL). Using flow cytometry, we observed increased NLRP3 and IL-1β expression in antigen-presenting cells from spleen, kidney, and aorta. We also observed increased IsoLG production in antigen-presenting cells from these organs. Primary culture of CD11c+ dendritic cells confirmed that low extracellular Mg2+ exerts a direct effect on these cells, stimulating IL-1β and IL-18 production. The present findings show that NLRP3 inflammasome activation and IsoLG-adduct formation are stimulated when dietary Mg2+ is depleted. Interventions and increased dietary Mg2+ consumption may prove beneficial in decreasing the prevalence of hypertension and cardiovascular disease.
Collapse
Affiliation(s)
- Ashley Pitzer Mutchler
- Vanderbilt University Department of Medicine, Division of Clinical Pharmacology, Nashville, TN, United States
| | - Linh Huynh
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Ritam Patel
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Tracey Lam
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Daniel Bain
- University of Pittsburgh Department of Geology, Pittsburgh, PA, United States
| | - Sydney Jamison
- Meharry Medical College Nashville, Nashville, TN, United States
| | - Annet Kirabo
- Vanderbilt University Department of Medicine, Division of Clinical Pharmacology, Nashville, TN, United States
| | - Evan C. Ray
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| |
Collapse
|
11
|
Imig JD. Bioactive lipids in hypertension. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2023; 97:1-35. [PMID: 37236756 PMCID: PMC10918458 DOI: 10.1016/bs.apha.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Hypertension is a major healthcare issue that afflicts one in every three adults worldwide and contributes to cardiovascular diseases, morbidity and mortality. Bioactive lipids contribute importantly to blood pressure regulation via actions on the vasculature, kidney, and inflammation. Vascular actions of bioactive lipids include blood pressure lowering vasodilation and blood pressure elevating vasoconstriction. Increased renin release by bioactive lipids in the kidney is pro-hypertensive whereas anti-hypertensive bioactive lipid actions result in increased sodium excretion. Bioactive lipids have pro-inflammatory and anti-inflammatory actions that increase or decrease reactive oxygen species and impact vascular and kidney function in hypertension. Human studies provide evidence that fatty acid metabolism and bioactive lipids contribute to sodium and blood pressure regulation in hypertension. Genetic changes identified in humans that impact arachidonic acid metabolism have been associated with hypertension. Arachidonic acid cyclooxygenase, lipoxygenase and cytochrome P450 metabolites have pro-hypertensive and anti-hypertensive actions. Omega-3 fish oil fatty acids eicosapentaenoic acid and docosahexaenoic acid are known to be anti-hypertensive and cardiovascular protective. Lastly, emerging fatty acid research areas include blood pressure regulation by isolevuglandins, nitrated fatty acids, and short chain fatty acids. Taken together, bioactive lipids are key contributors to blood pressure regulation and hypertension and their manipulation could decrease cardiovascular disease and associated morbidity and mortality.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
12
|
Power G, Padilla J. (Re)modeling high-salt diet-induced hypertension in mice. Am J Physiol Heart Circ Physiol 2023; 324:H470-H472. [PMID: 36827228 DOI: 10.1152/ajpheart.00093.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Gavin Power
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States.,NextGen Precision Health, University of Missouri, Columbia, Missouri, United States
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States.,NextGen Precision Health, University of Missouri, Columbia, Missouri, United States.,Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, United States
| |
Collapse
|
13
|
Abstract
Several clinical and large population studies indicate that women are more salt-sensitive than men, yet the precise mechanisms by which the sexually dimorphic onset manifests remains incompletely understood. Here, we evaluate recent epidemiological data and highlight current knowledge from studies investigating sex-specific mechanisms of salt-sensitive blood pressure (SSBP). Emerging evidence indicates that women of all ethnicities are more salt-sensitive than men, at all ages both premenopausal and postmenopausal. However, menopause exacerbates severity and prevalence of SSBP, suggesting that female sex chromosomes predispose to and female sex hormones mitigate SSBP. Results from both human and rodent studies support the contribution of enhanced and inappropriate activation of the aldosterone-ECMR (endothelial cell mineralocorticoid receptor) axis promoting vascular dysfunction in females. Increases in adrenal response to angiotensin II, in association with higher ECMR expression and activation of endothelial ENaC (epithelial sodium channel) in females compared to males, are emerging as central players in the development of endothelial dysfunction and SSBP in females. Female sex increases the prevalence and susceptibility of SSBP and sex hormones and sex chromosome complement may exert antagonistic effects in the development of the female heightened SSBP.
Collapse
Affiliation(s)
- Candee T. Barris
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jessica L. Faulkner
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Physiology Department, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric J. Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
14
|
Ertuglu LA, Laffer CL, Kirabo A. In Memoriam: Fernando Elijovich. Hypertension 2023; 80:e1-e3. [PMID: 36475860 DOI: 10.1161/hypertensionaha.122.20541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Lale A Ertuglu
- Division of Nephrology, Department of Medicine (L.A.E.), Vanderbilt University Medical Center, Nashville, TN
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Department of Medicine (C.L.L., A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine (C.L.L., A.K.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
15
|
Crorkin P, Hao S, Ferreri NR. Responses to Ang II (Angiotensin II), Salt Intake, and Lipopolysaccharide Reveal the Diverse Actions of TNF-α (Tumor Necrosis Factor-α) on Blood Pressure and Renal Function. Hypertension 2022; 79:2656-2670. [PMID: 36129177 PMCID: PMC9649876 DOI: 10.1161/hypertensionaha.122.19464] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
TNF-α (tumor necrosis factor-alpha) is the best known as a proinflammatory cytokine; yet, this cytokine also has important immunomodulatory and regulatory functions. As the effects of TNF-α on immune system function were being revealed, the spectrum of its activities appeared in conflict with each other before investigators defined the settings and mechanisms by which TNF-α contributed to both host defense and chronic inflammation. These effects reflect self-protective mechanisms that may become harmful when dysregulated. The paradigm of physiological and pathophysiological effects of TNF-α has since been uncovered in the lung, colon, and kidney where its role has been identified in pulmonary edema, electrolyte reabsorption, and blood pressure regulation, respectively. Recent studies on the prohypertensive and inflammatory effects of TNF-α in the cardiovascular system juxtaposed to those related to NaCl and blood pressure homeostasis, the response of the kidney to lipopolysaccharide, and protection against bacterial infections are helping define the mechanisms by which TNF-α modulates distinct functions within the kidney. This review discusses how production of TNF-α by renal epithelial cells may contribute to regulatory mechanisms that not only govern electrolyte excretion and blood pressure homeostasis but also maintain the appropriate local hypersalinity environment needed for optimizing the innate immune response to bacterial infections in the kidney. It is possible that the wide range of effects mediated by TNF-α may be related to severity of disease, amount of inflammation and TNF-α levels, and the specific cell types that produce this cytokine, areas that remain to be investigated further.
Collapse
Affiliation(s)
- Patrick Crorkin
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | - Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | | |
Collapse
|
16
|
Ferrario CM, Saha A, VonCannon JL, Meredith WJ, Ahmad S. Does the Naked Emperor Parable Apply to Current Perceptions of the Contribution of Renin Angiotensin System Inhibition in Hypertension? Curr Hypertens Rep 2022; 24:709-721. [PMID: 36272015 DOI: 10.1007/s11906-022-01229-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW To address contemporary hypertension challenges, a critical reexamination of therapeutic accomplishments using angiotensin converting enzyme inhibitors and angiotensin II receptor blockers, and a greater appreciation of evidence-based shortcomings from randomized clinical trials are fundamental in accelerating future progress. RECENT FINDINGS Medications targeting angiotensin II mechanism of action are essential for managing primary hypertension, type 2 diabetes, heart failure, and chronic kidney disease. While the ability of angiotensin converting enzyme inhibitors and angiotensin II receptor blockers to control blood pressure is undisputed, practitioners, hypertension specialists, and researchers hold low awareness of these drugs' limitations in preventing or reducing the risk of cardiovascular events. Biases in interpreting gained knowledge from data obtained in randomized clinical trials include a pervasive emphasis on using relative risk reduction over absolute risk reduction. Furthermore, recommendations for clinical practice in international hypertension guidelines fail to address the significance of a residual risk several orders of magnitude greater than the benefits. We analyze the limitations of the clinical trials that have led to current recommended treatment guidelines. We define and quantify the magnitude of the residual risk in published hypertension trials and explore how activation of alternate compensatory bioprocessing components within the renin angiotensin system bypass the ability of angiotensin converting enzyme inhibitors and angiotensin II receptor blockers to achieve a significant reduction in total and cardiovascular deaths. We complete this presentation by outlining the current incipient but promising potential of immunotherapy to block angiotensin II pathology alone or possibly in combination with other antihypertensive drugs. A full appreciation of the magnitude of the residual risk associated with current renin angiotensin system-based therapies constitutes a vital underpinning for seeking new molecular approaches to halt or even reverse the cardiovascular complications of primary hypertension and encourage investigating a new generation of ACE inhibitors and ARBs with increased capacity to reach the intracellular compartments at which Ang II can be generated.
Collapse
Affiliation(s)
- Carlos M Ferrario
- Laboratory of Translational Hypertension and Vascular Research, Department of General Surgery, Wake Forest School of Medicine, Medical Center Blvd, Atrium Health Wake Forest Baptist, Winston Salem, NC, 27157, USA.
| | - Amit Saha
- Department of Anesthesiology, Wake Forest School of Medicine, Medical Center Blvd, Atrium Health Wake Forest Baptist, Winston Salem, NC, 27157, USA
| | - Jessica L VonCannon
- Laboratory of Translational Hypertension and Vascular Research, Department of General Surgery, Wake Forest School of Medicine, Medical Center Blvd, Atrium Health Wake Forest Baptist, Winston Salem, NC, 27157, USA
| | - Wayne J Meredith
- Laboratory of Translational Hypertension and Vascular Research, Department of General Surgery, Wake Forest School of Medicine, Medical Center Blvd, Atrium Health Wake Forest Baptist, Winston Salem, NC, 27157, USA
| | - Sarfaraz Ahmad
- Laboratory of Translational Hypertension and Vascular Research, Department of General Surgery, Wake Forest School of Medicine, Medical Center Blvd, Atrium Health Wake Forest Baptist, Winston Salem, NC, 27157, USA
| |
Collapse
|
17
|
Pitzer A, Kleyman TR, Kirabo A. Kidney Tubular IL-1β ENaCtivation in Diabetes and Salt-Sensitive Hypertension. Circ Res 2022; 131:74-76. [PMID: 35737755 DOI: 10.1161/circresaha.122.321335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ashley Pitzer
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN (A.P., A.K.)
| | - Thomas R Kleyman
- Departments of Medicine, Cell Biology, Pharmacology and Chemical Biology, University of Pittsburgh, PA (T.R.K.)
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center Nashville, TN (A.P., A.K.)
| |
Collapse
|
18
|
Ilatovskaya DV, Levchenko V, Winsor K, Blass GR, Spires DR, Sarsenova E, Polina I, Zietara A, Paterson M, Kriegel AJ, Staruschenko A. Effects of elevation of ANP and its deficiency on cardiorenal function. JCI Insight 2022; 7:148682. [PMID: 35380994 PMCID: PMC9090260 DOI: 10.1172/jci.insight.148682] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Atrial natriuretic peptide (ANP), encoded by Nppa, is a vasodilatory hormone that promotes salt excretion. Genome-wide association studies identified Nppa as a causative factor of blood pressure development, and in humans, ANP levels were suggested as an indicator of salt sensitivity. This study aimed to provide insights into the effects of ANP on cardiorenal function in salt-sensitive hypertension. To address this question, hypertension was induced in SSNPPA-/- (knockout of Nppa in the Dahl Salt-Sensitive (SS) rat background) or SSWT (wild type Dahl SS) rats by a high salt diet challenge (HS, 4% NaCl for 21 days). Chronic infusion of ANP in SSWT rats attenuated the increase in blood pressure and cardiorenal damage. Overall, SSNPPA-/- strain demonstrated higher blood pressure and intensified cardiac fibrosis (with no changes in ejection fraction) compared to SSWT rats. Furthermore, SSNPPA-/- rats exhibited kidney hypertrophy and higher glomerular injury scores, reduced diuresis, and lower sodium and chloride excretion than SSWT when fed a HS diet. Additionally, the activity of epithelial Na+ channel (ENaC) was found to be increased in the collecting ducts of the SSNPPA-/- rats. Taken together, these data show promise for the therapeutic benefits of ANP and ANP-increasing drugs for treating salt-sensitive hypertension.
Collapse
Affiliation(s)
- Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta, United States of America
| | - Vladislav Levchenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, United States of America
| | - Kristen Winsor
- Department of Physiology, Medical College of Wisconsin, Milwaukee, United States of America
| | - Gregory R Blass
- Department of Physiology, Medical College of Wisconsin, Milwaukee, United States of America
| | - Denisha R Spires
- Department of Physiology, Medical College of Georgia, Augusta, United States of America
| | - Elizaveta Sarsenova
- Department of Medicine, Medical University of South Carolina, Charleston, United States of America
| | - Iuliia Polina
- Department of Medicine, Medical University of South Carolina, Charleston, United States of America
| | - Adrian Zietara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, United States of America
| | - Mark Paterson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, United States of America
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, United States of America
| | | |
Collapse
|
19
|
Wang X, Liu Y, Wang Y, Dong X, Wang Y, Yang X, Tian H, Li T. Protective Effect of Coriander ( Coriandrum sativum L.) on High-Fructose and High-Salt Diet-Induced Hypertension: Relevant to Improvement of Renal and Intestinal Function. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3730-3744. [PMID: 35315647 DOI: 10.1021/acs.jafc.2c00267] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hypertension has become a leading cardiovascular risk factor worldwide. In this study, we explored the salutary effects and relevant mechanisms of coriander (Coriandrum sativum L.), an herbal plant with culinary and medicinal values, on high-fructose and high-salt diet (HFSD)-induced hypertension in SD rats. Our results showed that oral administration of coriander (1.0 or 2.0 g/kg·bw) effectively attenuated HFSD-induced elevation of systolic blood pressure, diastolic blood pressure, and mean arterial pressure. Coriander also increased the serum levels of vasodilator factors (PGI2, NO, and eNOS), decreased Na+ retention and serum uric acid (UA) level, and ameliorated glucolipid profiles. qPCR results revealed that coriander downregulated the mRNA expression of NHE3, a Na+/H+ exchanger responsible for Na+ absorption, in kidney and small intestine. 16S rDNA sequencing showed that coriander altered the gut microbiota composition with the beneficial bacteria Bifidobacterium and Oscillibacter significantly enriched. Correlation analysis indicated that the abundance of Bifidobacterium was evidently correlated with levels of NHE3, NO, eNOS, and UA. LC-MS/MS analysis revealed that coriander contained a variety of flavonoids including rutin and quercetin. Conclusively, long-term consumption of coriander may ameliorate HFSD-induced hypertension by mitigating HFSD-caused abnormal changes in vascular endothelial function, renal and intestinal sodium absorption, glucolipid homeostasis, and gut microbiota in rats.
Collapse
Affiliation(s)
- Xiaoyuan Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yueyue Liu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yu Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xinyue Dong
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an 710119, Shaanxi, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Honglei Tian
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Ting Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| |
Collapse
|
20
|
Maiuolo J, Carresi C, Gliozzi M, Mollace R, Scarano F, Scicchitano M, Macrì R, Nucera S, Bosco F, Oppedisano F, Ruga S, Coppoletta AR, Guarnieri L, Cardamone A, Bava I, Musolino V, Paone S, Palma E, Mollace V. The Contribution of Gut Microbiota and Endothelial Dysfunction in the Development of Arterial Hypertension in Animal Models and in Humans. Int J Mol Sci 2022; 23:ijms23073698. [PMID: 35409057 PMCID: PMC8999124 DOI: 10.3390/ijms23073698] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
The maintenance of the physiological values of blood pressure is closely related to unchangeable factors (genetic predisposition or pathological alterations) but also to modifiable factors (dietary fat and salt, sedentary lifestyle, overweight, inappropriate combinations of drugs, alcohol abuse, smoking and use of psychogenic substances). Hypertension is usually characterized by the presence of a chronic increase in systemic blood pressure above the threshold value and is an important risk factor for cardiovascular disease, including myocardial infarction, stroke, micro- and macro-vascular diseases. Hypertension is closely related to functional changes in the endothelium, such as an altered production of vasoconstrictive and vasodilator substances, which lead to an increase in vascular resistance. These alterations make the endothelial tissue unresponsive to autocrine and paracrine stimuli, initially determining an adaptive response, which over time lead to an increase in risk or disease. The gut microbiota is composed of a highly diverse bacterial population of approximately 1014 bacteria. A balanced intestinal microbiota preserves the digestive and absorbent functions of the intestine, protecting from pathogens and toxic metabolites in the circulation and reducing the onset of various diseases. The gut microbiota has been shown to produce unique metabolites potentially important in the generation of hypertension and endothelial dysfunction. This review highlights the close connection between hypertension, endothelial dysfunction and gut microbiota.
Collapse
Affiliation(s)
- Jessica Maiuolo
- Laboratory of Pharmaceutical Biology, in IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy;
- Correspondence: (J.M.); (M.G.)
| | - Cristina Carresi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Micaela Gliozzi
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Correspondence: (J.M.); (M.G.)
| | - Rocco Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Federica Scarano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Miriam Scicchitano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Roberta Macrì
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Saverio Nucera
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Francesca Bosco
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Francesca Oppedisano
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Stefano Ruga
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Anna Rita Coppoletta
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Lorenza Guarnieri
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Antonio Cardamone
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
| | - Irene Bava
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Vincenzo Musolino
- Laboratory of Pharmaceutical Biology, in IRC-FSH Center, Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy;
| | - Sara Paone
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Ernesto Palma
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- Nutramed S.c.a.r.l, Complesso Ninì Barbieri, Roccelletta di Borgia, 88021 Catanzaro, Italy;
| | - Vincenzo Mollace
- IRC-FSH Department of Health Sciences, University “Magna Græcia” of Catanzaro Italy, 88021 Catanzaro, Italy; (C.C.); (R.M.); (F.S.); (M.S.); (R.M.); (S.N.); (F.B.); (F.O.); (S.R.); (A.R.C.); (L.G.); (A.C.); (I.B.); (E.P.); (V.M.)
- IRCCS San Raffaele, Via di Valcannuta 247, 00133 Rome, Italy
| |
Collapse
|
21
|
Borrelli S, De Nicola L, De Gregorio I, Polese L, Pennino L, Elefante C, Carbone A, Rappa T, Minutolo R, Garofalo C. Volume-Independent Sodium Toxicity in Peritoneal Dialysis: New Insights from Bench to Bed. Int J Mol Sci 2021; 22:ijms222312804. [PMID: 34884617 PMCID: PMC8657906 DOI: 10.3390/ijms222312804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 11/16/2022] Open
Abstract
Sodium overload is common in end-stage kidney disease (ESKD) and is associated with increased cardiovascular mortality that is traditionally considered a result of extracellular volume expansion. Recently, sodium storage was detected by Na23 magnetic resonance imaging in the interstitial tissue of the skin and other tissues. This amount of sodium is osmotically active, regulated by immune cells and the lymphatic system, escapes renal control, and, more importantly, is associated with salt-sensitive hypertension. In chronic kidney disease, the interstitial sodium storage increases as the glomerular filtration rate declines and is related to cardiovascular damage, regardless of the fluid overload. This sodium accumulation in the interstitial tissues becomes more significant in ESKD, especially in older and African American patients. The possible negative effects of interstitial sodium are still under study, though a higher sodium intake might induce abnormal structural and functional changes in the peritoneal wall. Interestingly, sodium stored in the interstial tissue is not unmodifiable, since it is removable by dialysis. Nevertheless, the sodium removal by peritoneal dialysis (PD) remains challenging, and new PD solutions are desirable. In this narrative review, we carried out an update on the pathophysiological mechanisms of volume-independent sodium toxicity and possible future strategies to improve sodium removal by PD.
Collapse
|