1
|
Navaneethabalakrishnan S, Goodlett B, Smith H, Cardenas A, Burns A, Mitchell B. Differential changes in end organ immune cells and inflammation in salt-sensitive hypertension: effects of lowering blood pressure. Clin Sci (Lond) 2024; 138:901-920. [PMID: 38949825 PMCID: PMC11250109 DOI: 10.1042/cs20240698] [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: 04/08/2024] [Revised: 06/17/2024] [Accepted: 07/01/2024] [Indexed: 07/02/2024]
Abstract
We reported that salt-sensitive hypertension (SSHTN) is associated with increased pro-inflammatory immune cells, inflammation, and inflammation-associated lymphangiogenesis in the kidneys and gonads of male and female mice. However, it is unknown whether these adverse end organ effects result from increased blood pressure (BP), elevated levels of salt, or both. We hypothesized that pharmaceutically lowering BP would not fully alleviate the renal and gonadal immune cell accumulation, inflammation, and lymphangiogenesis associated with SSHTN. SSHTN was induced in male and female C57BL6/J mice by administering nitro-L-arginine methyl ester hydrochloride (L-NAME; 0.5 mg/ml) in their drinking water for 2 weeks, followed by a 2-week washout period. Subsequently, the mice received a 3-week 4% high salt diet (SSHTN). The treatment group underwent the same SSHTN induction protocol but received hydralazine (HYD; 250 mg/L) in their drinking water during the diet phase (SSHTN+HYD). Control mice received tap water and a standard diet for 7 weeks. In addition to decreasing systolic BP, HYD treatment generally decreased pro-inflammatory immune cells and inflammation in the kidneys and gonads of SSHTN mice. Furthermore, the decrease in BP partially alleviated elevated renal and gonadal lymphatics and improved renal and gonadal function in mice with SSHTN. These data demonstrate that high systemic pressure and salt differentially act on end organ immune cells, contributing to the broader understanding of how BP and salt intake collectively shape immune responses and highlight implications for targeted therapeutic interventions.
Collapse
Affiliation(s)
| | - Bethany L. Goodlett
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Hannah L. Smith
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Alyssa Cardenas
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Asia Burns
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Brett M. Mitchell
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| |
Collapse
|
2
|
Navaneethabalakrishnan S, Goodlett B, Smith H, Montalvo R, Cardenas A, Mitchell B. Differential changes in end organ immune cells and inflammation in salt-sensitive hypertension: effects of increasing M2 macrophages. Clin Sci (Lond) 2024; 138:921-940. [PMID: 38949840 PMCID: PMC11250104 DOI: 10.1042/cs20240699] [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: 04/08/2024] [Revised: 06/18/2024] [Accepted: 07/01/2024] [Indexed: 07/02/2024]
Abstract
Salt-sensitive hypertension (SSHTN) is associated with M1 macrophage polarization and inflammatory responses, leading to inflammation-associated lymphangiogenesis and functional impairment across multiple organs, including kidneys and gonads. However, it remains unclear whether promoting M2 macrophage polarization can alleviate the hypertension, inflammation, and end organ damage in mice with salt sensitive hypertension (SSHTN). Male and female mice were made hypertensive by administering nitro-L-arginine methyl ester hydrochloride (L-NAME; 0.5 mg/ml) for 2 weeks in the drinking water, followed by a 2-week interval without any treatments, and a subsequent high salt diet for 3 weeks (SSHTN). AVE0991 (AVE) was intraperitoneally administered concurrently with the high salt diet. Control mice were provided standard diet and tap water. AVE treatment significantly attenuated BP and inflammation in mice with SSHTN. Notably, AVE promoted M2 macrophage polarization, decreased pro-inflammatory immune cell populations, and improved function in renal and gonadal tissues of mice with SSHTN. Additionally, AVE decreased lymphangiogenesis in the kidneys and testes of male SSHTN mice and the ovaries of female SSHTN mice. These findings highlight the effectiveness of AVE in mitigating SSHTN-induced elevated BP, inflammation, and end organ damage by promoting M2 macrophage polarization and suppressing pro-inflammatory immune responses. Targeting macrophage polarization emerges as a promising therapeutic approach for alleviating inflammation and organ damage in SSHTN. Further studies are warranted to elucidate the precise mechanisms underlying AVE-mediated effects and to assess its clinical potential in managing SSHTN.
Collapse
Affiliation(s)
| | - Bethany L. Goodlett
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Hannah L. Smith
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Robert A. Montalvo
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Alyssa Cardenas
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| | - Brett M. Mitchell
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX, U.S.A
| |
Collapse
|
3
|
Abstract
Salt (sodium chloride) is an essential nutrient required to maintain physiological functions. However, for most people, daily salt intake far exceeds their physiological need and is habitually greater than recommended upper thresholds. Excess salt intake leads to elevation in blood pressure which drives cardiovascular morbidity and mortality. Indeed, excessive salt intake is estimated to be responsible for ≈5 million deaths per year globally. For approximately one-third of otherwise healthy individuals (and >50% of those with hypertension), the effect of salt intake on blood pressure elevation is exaggerated; such people are categorized as salt sensitive and salt sensitivity of blood pressure is considered an independent risk factor for cardiovascular disease and death. The prevalence of salt sensitivity is higher in women than in men and, in both, increases with age. This narrative review considers the foundational concepts of salt sensitivity and the underlying effector systems that cause salt sensitivity. We also consider recent updates in preclinical and clinical research that are revealing new modifying factors that determine the blood pressure response to high salt intake.
Collapse
Affiliation(s)
- Matthew A Bailey
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom (M.A.B., N.D.)
| | - Neeraj Dhaun
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, United Kingdom (M.A.B., N.D.)
- Department of Renal Medicine, Royal Infirmary of Edinburgh, United Kingdom (N.D.)
| |
Collapse
|
4
|
Saleem M, Masenga SK, Ishimwe JA, Demirci M, Ahmad T, Jamison S, Albritton CF, Mwesigwa N, Porcia Haynes A, White J, Neikirk K, Vue Z, Hinton A, Arshad S, Desta S, Kirabo A. Recent Advances in Understanding Peripheral and Gut Immune Cell-Mediated Salt-Sensitive Hypertension and Nephropathy. Hypertension 2024; 81:436-446. [PMID: 38164753 PMCID: PMC10922672 DOI: 10.1161/hypertensionaha.123.22031] [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] [Indexed: 01/03/2024]
Abstract
Hypertension is the primary modifiable risk factor for cardiovascular, renal, and cerebrovascular diseases and is considered the main contributing factor to morbidity and mortality worldwide. Approximately 50% of hypertensive and 25% of normotensive people exhibit salt sensitivity of blood pressure, which is an independent risk factor for cardiovascular disease. Human and animal studies demonstrate that the immune system plays an important role in the etiology and pathogenesis of salt sensitivity of blood pressure, kidney damage, and vascular diseases. Antigen-presenting and adaptive immune cells are implicated in salt-sensitive hypertension and salt-induced renal and vascular injury. Elevated sodium activates antigen-presenting cells to release proinflammatory cytokines including IL (interleukin) 6, tumor necrosis factor-α, IL-1β, and accumulate isolevuglandin-protein adducts. In turn, these activate T cells release prohypertensive cytokines including IL-17A. Moreover, high-salt intake is associated with gut dysbiosis, leading to inflammation, oxidative stress, and blood pressure elevation but the mechanistic contribution to salt-sensitivity of blood pressure is not clearly understood. Here, we discuss recent advances in research investigating the cause, potential biomarkers, and therapeutic targets for salt-sensitive hypertension as they pertain to the gut microbiome, immunity, and inflammation.
Collapse
Affiliation(s)
- Mohammad Saleem
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sepiso K Masenga
- Mulungushi University, School of Medicine and Health Sciences, HAND Research Group, Livingstone, Zambia
| | - Jeanne A Ishimwe
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mert Demirci
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Taseer Ahmad
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Punjab, Pakistan
| | - Sydney Jamison
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN, USA
| | - Claude F. Albritton
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN, USA
| | - Naome Mwesigwa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandria Porcia Haynes
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jalyn White
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Spelman College Department of Chemistry and Biochemistry, Atlanta, GA, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Suha Arshad
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Selam Desta
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
5
|
Mannon EC, Muller PR, Sun J, Bush WB, Coleman A, Ocasio H, Polichnowski AJ, Brands MW, O'Connor PM. NaHCO3 loading causes increased arterial pressure and kidney damage in rats with chronic kidney disease. Clin Sci (Lond) 2024; 138:189-203. [PMID: 38300615 DOI: 10.1042/cs20231709] [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: 12/20/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/02/2024]
Abstract
Sodium bicarbonate (NaHCO3) is commonly utilized as a therapeutic to treat metabolic acidosis in people with chronic kidney disease (CKD). While increased dietary sodium chloride (NaCl) is known to promote volume retention and increase blood pressure, the effects of NaHCO3 loading on blood pressure and volume retention in CKD remain unclear. In the present study, we compared the effects of NaCl and NaHCO3 loading on volume retention, blood pressure, and kidney injury in both 2/3 and 5/6 nephrectomy remnant kidney rats, a well-established rodent model of CKD. We tested the hypothesis that NaCl loading promotes greater volume retention and increases in blood pressure than equimolar NaHCO3. Blood pressure was measured 24 h daily using radio telemetry. NaCl and NaHCO3 were administered in drinking water ad libitum or infused via indwelling catheters. Rats were housed in metabolic cages to determine volume retention. Our data indicate that both NaHCO3 and NaCl promote hypertension and volume retention in remnant kidney rats, with salt-sensitivity increasing with greater renal mass reduction. Importantly, while NaHCO3 intake was less pro-hypertensive than equimolar NaCl intake, NaHCO3 was not benign. NaHCO3 loading significantly elevated blood pressure and promoted volume retention in rats with CKD when compared with control rats receiving tap water. Our findings provide important insight into the effects of sodium loading with NaHCO3 in CKD and indicate that NaHCO3 loading in patients with CKD is unlikely to be benign.
Collapse
Affiliation(s)
- Elinor C Mannon
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - P Robinson Muller
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Jingping Sun
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Weston B Bush
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Alex Coleman
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Hiram Ocasio
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Aaron J Polichnowski
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, U.S.A
| | - Michael W Brands
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, U.S.A
| |
Collapse
|
6
|
Ou-Yang YN, Deng FF, Wang YJ, Chen M, Yang PF, Yang Z, Tian Z. High-salt diet induces dyslipidemia through the SREBP2/PCSK9 pathway in dahl salt-sensitive rats. Biochimie 2024; 216:34-45. [PMID: 37844755 DOI: 10.1016/j.biochi.2023.10.001] [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: 01/18/2023] [Revised: 09/24/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
A high-salt diet is known to increase serum cholesterol levels; however, the underlying mechanism of salt-induced dyslipidemia in patients with salt-sensitivity remains poorly understood. We aimed to investigate whether high-salt diet (HSD) can induce dyslipidemia and elucidate the underlying mechanism of salt-induced dyslipidemia in Dahl salt-sensitive (SS) rats. Metabolomic and biochemical analyses revealed that the consumption of an HSD (8 % NaCl) significantly increased the serum levels of total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) in SS rats. The enzyme-linked immunosorbent assay demonstrated an increase in circulating proprotein convertase subtilisin/kexin type 9 (PCSK9) levels, accompanied by a decrease in hepatic low-density lipoprotein receptor (LDLR) levels due to HSD consumption. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis revealed that HSD consumption activated sterol regulatory element-binding protein-2 (SREBP2) expression in the liver and kidney, resulting in upregulation of PCSK9 at the transcriptional level in the liver and at the translational level in the kidney, ultimately increasing circulating PCSK9 levels. The combined effects of HSD on the liver and kidney contributed to the development of hypercholesterolemia. Furthermore, an in vitro assay confirmed that high-salt exposure led to an increase in the protein expression of SREBP2 and PCSK9 secretion, thereby reducing low-density lipoprotein (LDL) uptake. This study, for the first time, shows that an HSD induces dyslipidemia through activation of the SREBP2/PCSK9 pathway, providing new insights into the prevention and treatment of dyslipidemia in patients with salt sensitivity.
Collapse
Affiliation(s)
- Ya-Nan Ou-Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Fen-Fen Deng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Yun-Jia Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Meng Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Peng-Fei Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Zhongmin Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| |
Collapse
|
7
|
Walton SD, Dasinger JH, Burns EC, Cherian-Shaw M, Abais-Battad JM, Mattson DL. Functional NADPH oxidase 2 in T cells amplifies salt-sensitive hypertension and associated renal damage. Am J Physiol Renal Physiol 2023; 325:F214-F223. [PMID: 37318993 PMCID: PMC10396224 DOI: 10.1152/ajprenal.00014.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/10/2023] [Accepted: 05/28/2023] [Indexed: 06/17/2023] Open
Abstract
Infiltrating T cells in the kidney amplify salt-sensitive (SS) hypertension and renal damage, but the mechanisms are not known. Genetic deletion of T cells (SSCD247-/-) or of the p67phox subunit of NADPH oxidase 2 (NOX2; SSp67phox-/-) attenuates SS hypertension in the Dahl SS rat. We hypothesized that reactive oxygen species produced by NOX2 in T cells drive the SS phenotype and renal damage. T cells were reconstituted by adoptively transferring splenocytes (∼10 million) from the Dahl SS (SS→CD247) rat, the SSp67phox-/- rat (p67phox→CD247), or only PBS (PBS→CD247) into the SSCD247-/- rat on postnatal day 5. Animals were instrumented with radiotelemeters and studied at 8 wk of age. There were no detectable differences in mean arterial pressure (MAP) or albuminuria between groups when rats were maintained on a low-salt (0.4% NaCl) diet. After 21 days of high-salt diet (4.0% NaCl), MAP and albuminuria were significantly greater in SS→CD247 rats compared with p67phox→CD247 and PBS→CD247 rats. Interestingly, there was no difference between p67phox→CD247 and PBS→CD247 rats in albuminuria or MAP after 21 days. The lack of CD3+ cells in PBS→CD247 rats and the presence of CD3+ cells in rats that received the T cell transfer demonstrated the effectiveness of the adoptive transfer. No differences in the number of CD3+, CD4+, or CD8+ cells were observed in the kidneys of SS→CD247 and p67phox→CD247 rats. These results indicate that reactive oxygen species produced by NOX2 in T cells participates in the amplification of SS hypertension and renal damage.NEW & NOTEWORTHY Our current work used the adoptive transfer of T cells that lack functional NADPH oxidase 2 into a genetically T cell-deficient Dahl salt-sensitive (SS) rat model. The results demonstrated that reactive oxygen species produced by NADPH oxidase 2 in T cells participate in the amplification of SS hypertension and associated renal damage and identifies a potential mechanism that exacerbates the salt-sensitive phenotype.
Collapse
Affiliation(s)
- Samuel D Walton
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - John Henry Dasinger
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Emily C Burns
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Mary Cherian-Shaw
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| | - David L Mattson
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
8
|
Poudel B, Ekperikpe US, Mandal S, Wilson GE, Shields CA, Cornelius DC, Williams JM. Chronic treatment with IL-25 increases renal M2 macrophages and reduces renal injury in obese Dahl salt-sensitive rats during the prepubescent stage. Am J Physiol Renal Physiol 2023; 325:F87-F98. [PMID: 37167270 PMCID: PMC10292980 DOI: 10.1152/ajprenal.00209.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/13/2023] Open
Abstract
Recently, we have reported that the early progression of proteinuria in the obese Dahl salt-sensitive (SS) leptin receptor mutant (SSLepRmutant) strain was associated with increased renal macrophage infiltration before puberty. Macrophages can be divided into two distinct phenotypes: M1 (proinflammatory) and M2 (anti-inflammatory). Moreover, previous studies have demonstrated that interleukin (IL)-25 converts resting macrophages and M1 into M2. Therefore, the present study examined whether treatment with IL-25 would reduce the early progression of renal injury in SSLepRmutant rats by increasing renal M2. We also investigated the impact of IL-25 on M2 subtypes: M2a (wound healing/anti-inflammatory), M2b (immune mediated/proinflammatory), M2c (regulatory/anti-inflammatory), and M2d (tumor associated/proangiogenic). Four-wk-old SS and SSLepRmutant rats were treated with either control (IgG) or IL-25 (1 µg/day ip every other day) for 4 wk. The kidneys from SSLepRmutant rats displayed progressive proteinuria and renal histopathology versus SS rats. IL-25 treatment had no effect on these parameters in SS rats. However, in the SSLepRmutant strain, proteinuria was markedly reduced after IL-25 treatment. Chronic treatment with IL-25 significantly decreased glomerular and tubular injury and renal fibrosis in the SSLepRmutant strain. Although the administration of IL-25 did not change total renal macrophage infiltration in both SS and SSLepRmutant rats, IL-25 increased M2a by >50% and reduced M1 by 60% in the kidneys of SSLepRmutant rats. Overall, these data indicate that IL-25 reduces the early progression of renal injury in SSLepRmutant rats by inducing M2a and suppressing M1 and suggest that IL-25 may be a therapeutic target for renal disease associated with obesity. NEW & NOTEWORTHY For the past few decades, immune cells and inflammatory cytokines have been demonstrated to play an important role in the development of renal disease. The present study provides strong evidence that interleukin-25 slows the early progression of renal injury in obese Dahl salt-sensitive rats before puberty by increasing systemic anti-inflammatory cytokines and renal M2a macrophages.
Collapse
Affiliation(s)
- Bibek Poudel
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Ubong S Ekperikpe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Sautan Mandal
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Gregory E Wilson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Corbin A Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| |
Collapse
|
9
|
Domingues da Silva CHN, Leite Guedes IH, de Lima JCS, Sobrinho JMDR, dos Santos AA. Responses Triggered by the Immune System in Hypertensive Conditions and Repercussions on Target Organ Damage: A Review. Curr Cardiol Rev 2023; 19:e200922208959. [PMID: 36125837 PMCID: PMC10201903 DOI: 10.2174/1573403x18666220920090632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/19/2022] [Accepted: 07/30/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Hypertension is a chronic, multifactorial clinical condition characterized by sustained high blood pressure levels. It is often associated with functional-structural alterations of target organs, which include heart, brain, kidneys, and vasculature. OBJECTIVE This study highlights the recent correlation between the immune system and hypertension and its repercussions on target-organ damage. METHODS The descriptors used for the search of the study were "hypertension", "immunity", and "target organs". The methodology of the study followed the main recommendations of the PRISMA statement. RESULTS The damage to the vasculature arises mainly from the migration of T cells and monocytes that become pro-inflammatory in the adventitia, releasing TNF-α, IFN-γ, and IL-17, which induce endothelial damage and hinder vascular relaxation. In the renal context, the inflammatory process associated with hypertension culminates in renal invasion by leukocytes, which contribute to the injury of this organ by mechanisms of intense sympathetic stimulation, activation of the reninangiotensin system, sodium retention, and aggravation of oxidative stress. In the cardiac context, hypertension increases the expression of pro-inflammatory elements, such as B, T, and NK cells, in addition to the secretion of IFN-γ, IL-17, IL-23, and TNF-α from angiotensin II, reactive oxygen species, and aldosterone. This pro-inflammatory action is also involved in brain damage through SphK1. In view of the above, the participation of the immune system in hypertension-induced injuries seems to be unequivocal. CONCLUSION Therefore, understanding the multifactorial mechanisms related to hypertension will certainly allow for more efficient interventions in this condition, preventing target organ damage.
Collapse
Affiliation(s)
| | | | | | | | - Angela Amancio dos Santos
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pernambuco, Joao Pessoa 58051-085, Brazil
| |
Collapse
|
10
|
Zietara A, Spires DR, Juffre A, Costello HM, Crislip GR, Douma LG, Levchenko V, Dissanayake LV, Klemens CA, Nikolaienko O, Geurts AM, Gumz ML, Staruschenko A. Knockout of the Circadian Clock Protein PER1 (Period1) Exacerbates Hypertension and Increases Kidney Injury in Dahl Salt-Sensitive Rats. Hypertension 2022; 79:2519-2529. [PMID: 36093781 PMCID: PMC9669134 DOI: 10.1161/hypertensionaha.122.19316] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Circadian rhythms play an essential role in physiological function. The molecular clock that underlies circadian physiological function consists of a core group of transcription factors, including the protein PER1 (Period1). Studies in mice show that PER1 plays a role in the regulation of blood pressure and renal sodium handling; however, the results are dependent on the strain being studied. Using male Dahl salt-sensitive (SS) rats with global knockout of PER1 (SSPer1-/-), we aim to test the hypothesis that PER1 plays a key role in the regulation of salt-sensitive blood pressure. METHODS The model was generated using CRISPR/Cas9 and was characterized using radiotelemetry and measures of renal function and circadian rhythm. RESULTS SSPer1-/- rats had similar mean arterial pressure when fed a normal 0.4% NaCl diet but developed augmented hypertension after three weeks on a high-salt (4% NaCl) diet. Despite being maintained on a normal 12:12 light:dark cycle, SSPer1-/- rats exhibited desynchrony mean arterial pressure rhythms on a high-salt diet, as evidenced by increased variability in the time of peak mean arterial pressure. SSPer1-/- rats excrete less sodium after three weeks on the high-salt diet. Furthermore, SSPer1-/- rats exhibited decreased creatinine clearance, a measurement of renal function, as well as increased signs of kidney tissue damage. SSPer1-/- rats also exhibited higher plasma aldosterone levels. CONCLUSIONS Altogether, our findings demonstrate that loss of PER1 in Dahl SS rats causes an array of deleterious effects, including exacerbation of the development of salt-sensitive hypertension and renal damage.
Collapse
Affiliation(s)
- Adrian Zietara
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Denisha R. Spires
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Alexandria Juffre
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Hannah M. Costello
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - G. Ryan Crislip
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
| | - Lauren G. Douma
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
| | - Vladislav Levchenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
| | - Lashodya V. Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
| | - Christine A. Klemens
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33602, USA
| | - Oksana Nikolaienko
- Department of Cellular Membranology, Bogomoletz Institute of Physiology, Kyiv, Ukraine
| | - Aron M. Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Michelle L. Gumz
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL 32610, USA
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32610, USA
- Department of Medicine, Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL 32610, USA
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL 32610, USA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33602, USA
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL 33602, USA
- James A. Haley Veterans’ Hospital, Tampa, FL 33612, USA
| |
Collapse
|
11
|
Navaneethabalakrishnan S, Smith HL, Arenaz CM, Goodlett BL, McDermott JG, Mitchell BM. Update on Immune Mechanisms in Hypertension. Am J Hypertens 2022; 35:842-851. [PMID: 35704473 PMCID: PMC9527774 DOI: 10.1093/ajh/hpac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/02/2023] Open
Abstract
The contribution of immune cells in the initiation and maintenance of hypertension is undeniable. Several studies have established the association between hypertension, inflammation, and immune cells from the innate and adaptive immune systems. Here, we provide an update to our 2017 American Journal of Hypertension review on the overview of the cellular immune responses involved in hypertension. Further, we discuss the activation of immune cells and their contribution to the pathogenesis of hypertension in different in vivo models. We also highlight existing gaps in the field of hypertension that need attention. The main goal of this review is to provide a knowledge base for translational research to develop therapeutic strategies that can improve cardiovascular health in humans.
Collapse
Affiliation(s)
| | - Hannah L Smith
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Cristina M Arenaz
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Bethany L Goodlett
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Justin G McDermott
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M College of Medicine, Bryan, Texas, USA
| |
Collapse
|
12
|
Li X, Alu A, Wei Y, Wei X, Luo M. The modulatory effect of high salt on immune cells and related diseases. Cell Prolif 2022; 55:e13250. [PMID: 35747936 PMCID: PMC9436908 DOI: 10.1111/cpr.13250] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The adverse effect of excessive salt intake has been recognized in decades. Researchers have mainly focused on the association between salt intake and hypertension. However, studies in recent years have proposed the existence of extra-renal sodium storage and provided insight into the immunomodulatory function of sodium. OBJECTIVES In this review, we discuss the modulatory effects of high salt on various innate and adaptive immune cells and immune-regulated diseases. METHODS We identified papers through electronic searches of PubMed database from inception to March 2022. RESULTS An increasing body of evidence has demonstrated that high salt can modulate the differentiation, activation and function of multiple immune cells. Furthermore, a high-salt diet can increase tissue sodium concentrations and influence the immune responses in microenvironments, thereby affecting the development of immune-regulated diseases, including hypertension, multiple sclerosis, cancer and infections. These findings provide a novel mechanism for the pathology of certain diseases and indicate that salt might serve as a target or potential therapeutic agent in different disease contexts. CONCLUSION High salt has a profound impact on the differentiation, activation and function of multiple immune cells. Additionally, an HSD can modulate the development of various immune-regulated diseases.
Collapse
Affiliation(s)
- Xian Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Min Luo
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Hypertension Induces Gonadal Macrophage Imbalance, Inflammation, Lymphangiogenesis, and Dysfunction. Clin Sci (Lond) 2022; 136:879-894. [PMID: 35532133 DOI: 10.1042/cs20220117] [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: 02/16/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/17/2022]
Abstract
Hypertension (HTN) is associated with gonadal dysfunction and impaired reproductive health in both men and women. An imbalance in the systemic and renal pro-inflammatory (M1)/anti-inflammatory (M2) macrophage ratio, increased inflammation, and inflammation-associated lymphangiogenesis have been observed in animals with HTN. However, the impact of HTN on gonadal macrophages, inflammation, and lymphatics remains obscure. We hypothesized that salt-sensitive HTN (SSHTN) and HTN alters gonadal macrophage polarization, which is associated with inflammation, inflammation-associated lymphangiogenesis and reproductive dysfunction. Flow cytometry analyses revealed a significant increase in M1 macrophages in the testes of SSHTN and nitric oxide synthase inhibition-induced HTN (LHTN) mice, with a concurrent decrease in M2 macrophages in SSHTN mice yet an increase in M2 macrophages in LHTN mice. Ovaries from SSHTN mice exhibited increase in M1 and a decrease in M2 macrophages, while ovaries from LHTN mice had a significant increase in M2 and a decrease in M1 macrophages. Gene expression patterns of pro-inflammatory cytokines revealed gonadal inflammation in all hypertensive mice. Increased lymphatic vessel density in the gonads of both male and female hypertensive mice was confirmed by immunofluorescence staining for LYVE-1. HTN adversely affected the expression pattern of steroidogenic enzymes, hormone receptors, and secretory proteins in both the testes and ovaries. In line with these results, male hypertensive mice also presented with decreased sperm concentration, and increased percentage of sperm with abnormal morphology, damaged acrosome, and non-functional mitochondrial activity. These data demonstrate that HTN alters gonadal macrophage polarization, which is associated with gonadal inflammation, inflammation-associated lymphangiogenesis, and dysfunction.
Collapse
|
14
|
Poudel B, Shields CA, Ekperikpe US, Brown AK, Travis OK, Maury JC, Fitzgerald S, Smith SV, Cornelius DC, Williams JM. The SS LepR mutant rat represents a novel model to study obesity-induced renal injury before puberty. Am J Physiol Regul Integr Comp Physiol 2022; 322:R299-R308. [PMID: 35107024 PMCID: PMC8917907 DOI: 10.1152/ajpregu.00179.2021] [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] [Received: 07/16/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 02/08/2023]
Abstract
Prepubertal obesity (PPO) has emerged as a major health problem over the past few decades and is a risk factor for the development of proteinuria. The current study investigated whether the development of renal injury in the obese SSLepR mutant strain occurs before puberty. When determining the temporal changes in serum sex hormones in female and male SS and SSLepR mutant rats between 4 and 10 wk of age, we only observed significant increases in estradiol and testosterone levels in female and male SS rats at 10 wk of age than at 4 wk of age. The results suggest that studying both strains between 4 and 8 wk of age is appropriate to study the effects of PPO on renal injury in this model. Proteinuria was significantly higher in SSLepR mutant rats as opposed to the values observed in SS rats at 8 wk of age, and we did not observe any sex differences in proteinuria in either strain. The kidneys from the SSLepR mutant rats displayed significant glomerular and tubular injury and renal fibrosis versus the values measured in SS rats without any sex differences. Overall, we observed increased immune cell infiltration in the kidneys from SSLepR mutant rats compared with SS rats. Interestingly, female SSLepR mutant rats displayed significant increases in not only M1 macrophages (proinflammatory) but also M2 macrophages (anti-inflammatory) versus male SSLepR mutant rats. These results suggest the SSLepR mutant rat may be a useful model to study early progression of obesity-related renal injury before the onset of puberty.
Collapse
Affiliation(s)
- Bibek Poudel
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Corbin A Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ubong S Ekperikpe
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Andrea K Brown
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Olivia K Travis
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jordan C Maury
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Sarah Fitzgerald
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Stanley V Smith
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
15
|
Sylvester MA, Pollow DP, Moffett C, Nunez W, Uhrlaub JL, Nikolich-Zugich J, Brooks HL. Splenocyte transfer from hypertensive donors eliminates premenopausal female protection from ANG II-induced hypertension. Am J Physiol Renal Physiol 2022; 322:F245-F257. [PMID: 35001661 PMCID: PMC8858666 DOI: 10.1152/ajprenal.00369.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/15/2021] [Accepted: 01/04/2022] [Indexed: 11/22/2022] Open
Abstract
Premenopausal females are protected from angiotensin II (ANG II)-induced hypertension following the adoptive transfer of T cells from normotensive donors. For the present study, we hypothesized that the transfer of hypertensive T cells (HT) or splenocytes (HS) from hypertensive donors would eliminate premenopausal protection from hypertension. Premenopausal recombination-activating gene-1 (Rag-1)-/- females received either normotensive (NT) or hypertensive cells 3 wk before ANG II infusion (14 days, 490 ng/kg/min). Contrary to our hypothesis, no increase in ANG II-induced blood pressure was observed in the NT/ANG or HT/ANG groups. Flow cytometry demonstrated that renal FoxP3+ T regulatory cells were significantly decreased, and immunohistochemistry showed an increase in renal F4/80+ macrophages in the HT/ANG group, suggesting a shift in the renal inflammatory environment despite no change in blood pressure. Renal mRNA expression of macrophage chemoattractant protein-1 (MCP-1), endothelin-1 (ET-1), and G protein-coupled estrogen receptor-1 (GPER-1) was significantly decreased in the HT/ANG group. The adoptive transfer of hypertensive splenocytes before ANG II infusion (HS/ANG) eliminated premenopausal protection from hypertension and significantly decreased splenic FoxP3+ T regulatory cells compared with females that received normotensive splenocytes (NS/ANG). Expression of macrophage inflammatory protein 1α/chemokine (C-C motif) ligand 3 (MCP-1/CCL3), a potent macrophage chemokine, was elevated in the HS/ANG group; however, no increase in renal macrophage infiltration occurred. Together, these data show that in premenopausal females, T cells from hypertensive donors are not sufficient to induce robust ANG II-mediated hypertension; in contrast, transfer of hypertensive splenocytes (consisting of T/B lymphocytes, dendritic cells, and macrophages) is sufficient. Further work is needed to understand how innate and adaptive immune cells and estrogen signaling coordinate to cause differential hypertensive outcomes in premenopausal females.NEW & NOTEWORTHY Our study is the first to explore the role of hypertensive T cells versus hypertensive splenocytes in premenopausal protection from ANG II-induced hypertension. We show that the hypertensive status of T cell donors does not impact blood pressure in the recipient female. However, splenocytes, when transferred from hypertensive donors, significantly increased premenopausal recipient blood pressure following ANG II infusion, highlighting the importance of further investigation into estrogen signaling and immune cell activation in females.
Collapse
Affiliation(s)
| | - Dennis P Pollow
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Caitlin Moffett
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Wendy Nunez
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona, Tucson, Arizona
- University of Arizona Center on Aging, University of Arizona, Tucson, Arizona
| | - Janko Nikolich-Zugich
- Department of Immunobiology, University of Arizona, Tucson, Arizona
- University of Arizona Center on Aging, University of Arizona, Tucson, Arizona
| | - Heddwen L Brooks
- Department of Physiology, University of Arizona, Tucson, Arizona
- Sarver Heart Center, University of Arizona, Tucson, Arizona
- University of Arizona Center on Aging, University of Arizona, Tucson, Arizona
| |
Collapse
|
16
|
Tuong ZK, Stewart BJ, Guo SA, Clatworthy MR. Epigenetics and tissue immunity-Translating environmental cues into functional adaptations. Immunol Rev 2021; 305:111-136. [PMID: 34821397 DOI: 10.1111/imr.13036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022]
Abstract
There is an increasing appreciation that many innate and adaptive immune cell subsets permanently reside within non-lymphoid organs, playing a critical role in tissue homeostasis and defense. The best characterized are macrophages and tissue-resident T lymphocytes that work in concert with organ structural cells to generate appropriate immune responses and are functionally shaped by organ-specific environmental cues. The interaction of tissue epithelial, endothelial and stromal cells is also required to attract, differentiate, polarize and maintain organ immune cells in their tissue niche. All of these processes require dynamic regulation of cellular transcriptional programmes, with epigenetic mechanisms playing a critical role, including DNA methylation and post-translational histone modifications. A failure to appropriately regulate immune cell transcription inevitably results in inadequate or inappropriate immune responses and organ pathology. Here, with a focus on the mammalian kidney, an organ which generates differing regional environmental cues (including hypersalinity and hypoxia) due to its physiological functions, we will review the basic concepts of tissue immunity, discuss the technologies available to profile epigenetic modifications in tissue immune cells, including those that enable single-cell profiling, and consider how these mechanisms influence the development, phenotype, activation and function of different tissue immune cell subsets, as well as the immunological function of structural cells.
Collapse
Affiliation(s)
- Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Shuang Andrew Guo
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK.,Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| |
Collapse
|
17
|
Innate immunity and clinical hypertension. J Hum Hypertens 2021; 36:503-509. [PMID: 34689174 DOI: 10.1038/s41371-021-00627-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 01/10/2023]
Abstract
Emerging evidence has supported a role of inflammation and immunity in the genesis of hypertension. In humans and experimental models of hypertension, cells of the innate and adaptive immune system enter target tissues, including vessels and the kidney, and release powerful mediators including cytokines, matrix metalloproteinases and reactive oxygen species that cause tissue damage, fibrosis and dysfunction. These events augment the blood pressure elevations in hypertension and promote end-organ damage. Factors that activate immune cells include sympathetic outflow, increased sodium within microenvironments where these cells reside, and signals received from the vasculature. In particular, the activated endothelium releases reactive oxygen species and interleukin (IL)-6 which in turn stimulate transformation of monocytes to become antigen presenting cells and produce cytokines like IL-1β and IL-23, which further affect T cell function to produce IL-17A. Genetic deletion or neutralization of these cytokines ameliorates hypertension and end-organ damage. In this review, we will consider in depth features of the hypertensive milieu that lead to these events and consider new treatment approaches to limit the untoward effects of inflammation in hypertension.
Collapse
|
18
|
Olorunnisola OS, Adegbola PI, Ajilore BS, Akintola OA, Fadahunsi OS. The Role of Poly-Herbal Extract in Sodium Chloride-Induced Oxidative Stress and Hyperlipidemia in Male Wistar Rats. MEDICINES (BASEL, SWITZERLAND) 2021; 8:medicines8060025. [PMID: 34072736 PMCID: PMC8228643 DOI: 10.3390/medicines8060025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/11/2021] [Accepted: 05/28/2021] [Indexed: 05/06/2023]
Abstract
Consistent consumption of high salt diet (HSD) has been associated with increased cellular generation of free radicals, which has been implicated in the derangement of some vital organs and etiology of cardiovascular disorders. This study was designed to investigate the combined effect of some commonly employed medicinal plants on serum lipid profile and antioxidant status of aorta, kidney, and liver of high salt diet-fed animals. Out of the total fifty male Wistar rats obtained, fifteen were used for acute toxicity study, while the remaining thirty-five were divided into 5 groups of 7 animals each. Group 1 and 2 animals were fed normal rat chow (NRC) and 16% high salt diet (HSD) only, respectively. Animals in groups 3, 4 and 5 were fed 16% HSD with 800, 400, and 200 mg/kg bw poly-herbal extract (PHE), respectively, once for 28 consecutive days. Serum low-density lipoprotein (LDL), triacylglycerol (TG), total cholesterol (TC) and high-density lipoprotein (HDL), malondialdehyde, nitric oxide, catalase, superoxide dismutase, glutathione peroxidase, glutathione concentration, and activities were assessed in the aorta, kidney, and liver. Poly-herbal extract (p < 0.05) significantly reduced malondialdehyde and nitric oxide concentrations and also increased antioxidant enzymes and glutathione activity. Elevated serum TG, TC, LDL, and TC content in HSD-fed animals were significantly (p < 0.05) reduced to normal in PHE-treated rats while HDL was significantly elevated (p < 0.05) in a concentration-dependent manner in PHE treated animals. Feeding with PHE attenuated high-salt diet imposed derangement in serum lipid profile and antioxidant status in the organs of the experimental rats.
Collapse
Affiliation(s)
- Olubukola Sinbad Olorunnisola
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso P.M.B 4000, Oyo State, Nigeria; (O.S.O.); (P.I.A.)
| | - Peter Ifeoluwa Adegbola
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso P.M.B 4000, Oyo State, Nigeria; (O.S.O.); (P.I.A.)
| | - Bamidele Stephen Ajilore
- Department of Medical Biochemistry, Osun State University, Osogbo P.M.B 4494, Osun State, Nigeria;
| | - Olayemi Adebola Akintola
- Department of Science Laboratory and Technology, Faculty of Pure of Applied Sciences, Ladoke Akintola University of Technology, Ogbomoso P.M.B 4000, Oyo State, Nigeria;
| | - Olumide Samuel Fadahunsi
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso P.M.B 4000, Oyo State, Nigeria; (O.S.O.); (P.I.A.)
- Correspondence:
| |
Collapse
|
19
|
Wenstedt EFE, van Croonenburg TJ, van den Born BJH, Van den Bossche J, Hooijmans CR, Vogt L. The effect of macrophage-targeted interventions on blood pressure - a systematic review and meta-analysis of preclinical studies. Transl Res 2021; 230:123-138. [PMID: 33166696 DOI: 10.1016/j.trsl.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/08/2020] [Accepted: 11/03/2020] [Indexed: 01/01/2023]
Abstract
An increasing body of evidence shows a role for macrophages and monocytes (as their precursors) in hypertension, but with conflicting results with regard to whether they are protective or harmful. Therefore, we systematically reviewed the effect of macrophage interventions on blood pressure in animal models, to explore which factors determine the blood pressure increasing vs. decreasing effect. A search in PubMED and EMBASE yielded 9620 records, 26 of which were included. Eighteen studies (involving 22 different experiments (k = 22)) performed macrophage depletion, whereas 12 studies specifically deleted certain macrophage proteins. The blood pressure effects of macrophage depletion were highly various and directed toward both directions, as expected, which could not be reduced to differences in animal species or methods of hypertension induction. Prespecified subgroup analysis did reveal a potential role for the route in which the macrophage-depleting agent is being administrated (intraperitoneal vs intravenous subgroup difference of P = 0.07 (k = 22), or P < 0.001 in studies achieving considerable (ie, >50%) depletion (k = 18)). Along with findings from specific macrophage protein deletion studies-showing that deletion of one single macrophage protein (like TonEBP, endothelin-B, EP4, NOX-2 and the angiotensin II type 1 receptor) can alter blood pressure responses to hypertensive stimuli-the indication that each route has its specific depletion pattern regarding targeted tissues and macrophage phenotypes suggests a determinative role for these features. These hypothesis-generating results encourage more detailed depletion characterization of each technique by direct experimental comparisons, providing a chance to obtain more knowledge on which macrophages are beneficial versus detrimental in hypertension development.
Collapse
Affiliation(s)
- Eliane F E Wenstedt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Thirza J van Croonenburg
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Bert-Jan H van den Born
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Jan Van den Bossche
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Carlijn R Hooijmans
- Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE), Department of Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Liffert Vogt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Mattson DL, Dasinger JH, Abais-Battad JM. Amplification of Salt-Sensitive Hypertension and Kidney Damage by Immune Mechanisms. Am J Hypertens 2021; 34:3-14. [PMID: 32725162 PMCID: PMC7891248 DOI: 10.1093/ajh/hpaa124] [Citation(s) in RCA: 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
|
21
|
Liu Y, Dai X, Yang S, Peng Y, Hou F, Zhou Q. High salt aggravates renal inflammation via promoting pro-inflammatory macrophage in 5/6-nephrectomized rat. Life Sci 2021; 274:119109. [PMID: 33513393 DOI: 10.1016/j.lfs.2021.119109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/11/2021] [Accepted: 01/17/2021] [Indexed: 12/22/2022]
Abstract
The increasing incident of chronic kidney disease (CKD) in recent years might be related to a change in dietary habits, known as excessive salt intake. Given excessive salt promotes pathogenic T cells responses. Since the importance of macrophage in the development of CKD, we addressed the effect of high salt loading on in a rat CKD model. We observed that 5/6Nx rats receiving a high salt diet showed strongly enhanced macrophage infiltration and activation in the renal tissue accompanied by deteriorated renal inflammation. Then we used the microarray expression profiling to detect the effect of additional Nacl on peritoneal macrophage derived from 5/6Nx. The NaCl treatment of macrophage extracted from 5/6Nx rat elicited a strong pro-inflammatory phenotype characterized by enhanced proinflammatory cytokine production, increased expression of molecules mainly involved in immune response process. This NaCl-induced pro-inflammatory macrophage phenotype was accompanied by increased phosphorylation of STAT1. Taken together, our study demonstrated that high salt intake promotes immune activation of macrophages through the STAT1 independently and exacerbates the kidney accompanied by promotion of inflammation. Thus, changes in diet may provide a novel strategy for the prevention or amelioration of CKD.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyan Dai
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaohua Yang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Peng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fanfan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiugen Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
22
|
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
|
23
|
Fehrenbach DJ, Abais-Battad JM, Dasinger JH, Lund H, Keppel T, Zemaj J, Cherian-Shaw M, Gundry RL, Geurts AM, Dwinell MR, Mattson DL. Sexual Dimorphic Role of CD14 (Cluster of Differentiation 14) in Salt-Sensitive Hypertension and Renal Injury. Hypertension 2020; 77:228-240. [PMID: 33249861 DOI: 10.1161/hypertensionaha.120.14928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Genomic sequence and gene expression association studies in animals and humans have identified genes that may be integral in the pathogenesis of various diseases. CD14 (cluster of differentiation 14)-a cell surface protein involved in innate immune system activation-is one such gene associated with cardiovascular and hypertensive disease. We previously showed that this gene is upregulated in renal macrophages of Dahl salt-sensitive animals fed a high-salt diet; here we test the hypothesis that CD14 contributes to the elevated pressure and renal injury observed in salt-sensitive hypertension. Using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9), we created a targeted mutation in the CD14 gene on the Dahl SS (SS/JrHSDMcwi) background and validated the absence of CD14 peptides via mass spectrometry. Radiotelemetry was used to monitor blood pressure in wild-type and CD14-/- animals challenged with high salt and identified infiltrating renal immune cells via flow cytometry. Germline knockout of CD14 exacerbated salt-sensitive hypertension and renal injury in female animals but not males. CD14-/- females demonstrated increased infiltrating macrophages but no difference in infiltrating lymphocytes. Transplant of CD14+/+ or CD14-/- bone marrow was used to isolate the effects of CD14 knockout to hematopoietic cells and confirmed that the differential phenotype observed was due to knockout of CD14 in hematopoietic cells. Ovariectomy was used to remove the influence of female sex hormones, which completely abrogated the effect of CD14 knockout. These studies provide a novel treatment target and evidence of a new dichotomy in immune activation between sexes within the context of hypertensive disease where CD14 regulates immune cell activation and renal injury.
Collapse
Affiliation(s)
- Daniel J Fehrenbach
- Department of Physiology (D.J.F., H.L., J.Z., A.M.G., M.R.D.), Medical College of Wisconsin, Wauwatosa, WI.,Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, GA (D.J.F., J.M.A.-B., J.H.D., M.C.-S., D.L.M.)
| | - Justine M Abais-Battad
- Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, GA (D.J.F., J.M.A.-B., J.H.D., M.C.-S., D.L.M.)
| | - John Henry Dasinger
- Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, GA (D.J.F., J.M.A.-B., J.H.D., M.C.-S., D.L.M.)
| | - Hayley Lund
- Department of Physiology (D.J.F., H.L., J.Z., A.M.G., M.R.D.), Medical College of Wisconsin, Wauwatosa, WI
| | - Theodore Keppel
- Center for Biomedical Mass Spectrometry Research (T.K., R.L.G.), Medical College of Wisconsin, Wauwatosa, WI
| | - Jeylan Zemaj
- Department of Physiology (D.J.F., H.L., J.Z., A.M.G., M.R.D.), Medical College of Wisconsin, Wauwatosa, WI
| | - Mary Cherian-Shaw
- Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, GA (D.J.F., J.M.A.-B., J.H.D., M.C.-S., D.L.M.)
| | - Rebekah L Gundry
- Center for Biomedical Mass Spectrometry Research (T.K., R.L.G.), Medical College of Wisconsin, Wauwatosa, WI.,CardiOmics Program, Center for Heart and Vascular Research (R.L.G.), University of Nebraska Medical Center, Omaha, NE.,Division of Cardiovascular Medicine (R.L.G.), University of Nebraska Medical Center, Omaha, NE.,Department of Cellular and Integrative Physiology (R.L.G.), University of Nebraska Medical Center, Omaha, NE
| | - Aron M Geurts
- Department of Physiology (D.J.F., H.L., J.Z., A.M.G., M.R.D.), Medical College of Wisconsin, Wauwatosa, WI.,Genomic Sciences and Precision Medicine Center (A.M.G., M.R.D.), Medical College of Wisconsin, Wauwatosa, WI
| | - Melinda R Dwinell
- Department of Physiology (D.J.F., H.L., J.Z., A.M.G., M.R.D.), Medical College of Wisconsin, Wauwatosa, WI.,Genomic Sciences and Precision Medicine Center (A.M.G., M.R.D.), Medical College of Wisconsin, Wauwatosa, WI
| | - David L Mattson
- Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, GA (D.J.F., J.M.A.-B., J.H.D., M.C.-S., D.L.M.)
| |
Collapse
|
24
|
Klemens CA, Staruschenko A. American Journal of Physiology-Renal Physiology Collections: Hypertension. Am J Physiol Renal Physiol 2020; 319:F1001-F1002. [PMID: 33166184 DOI: 10.1152/ajprenal.00572.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Christine A Klemens
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin
| |
Collapse
|
25
|
Dey AB, Khedr S, Bean J, Porras LL, Meredith TD, Willard FS, Hass JV, Zhou X, Terashvili M, Jesudason CD, Ruley KM, Wiley MR, Kowala M, Atkinson SJ, Staruschenko A, Rekhter MD. Selective Phosphodiesterase 1 Inhibitor BTTQ Reduces Blood Pressure in Spontaneously Hypertensive and Dahl Salt Sensitive Rats: Role of Peripheral Vasodilation. Front Physiol 2020; 11:543727. [PMID: 33013477 PMCID: PMC7506137 DOI: 10.3389/fphys.2020.543727] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 08/14/2020] [Indexed: 01/20/2023] Open
Abstract
Regulation of the peripheral vascular resistance via modulating the vessel diameter has been considered as a main determinant of the arterial blood pressure. Phosphodiesterase enzymes (PDE1-11) hydrolyse cyclic nucleotides, which are key players controlling the vessel diameter and, thus, peripheral resistance. Here, we have tested and reported the effects of a novel selective PDE1 inhibitor (BTTQ) on the cardiovascular system. Normal Sprague Dawley, spontaneously hypertensive (SHR), and Dahl salt-sensitive rats were used to test in vivo the efficacy of the compound. Phosphodiesterase radiometric enzyme assay revealed that BTTQ inhibited all three isoforms of PDE1 in nanomolar concentration, while micromolar concentrations were needed to induce effective inhibition for other PDEs. The myography study conducted on mesenteric arteries revealed a potent vasodilatory effect of the drug, which was confirmed in vivo by an increase in the blood flow in the rat ear arteriols reflected by the rise in the temperature. Furthermore, BTTQ proved a high efficacy in lowering the blood pressure about 9, 36, and 24 mmHg in normal Sprague Dawley, SHR and, Dahl salt-sensitive rats, respectively, compared to the vehicle-treated group. Moreover, additional blood pressure lowering of about 22 mmHg could be achieved when BTTQ was administered on top of ACE inhibitor lisinopril, a current standard of care in the treatment of hypertension. Therefore, PDE1 inhibition induced efficient vasodilation that was accompanied by a significant reduction of blood pressure in different hypertensive rat models. Administration of BTTQ was also associated with increased heart rate in both models of hypertension as well as in the normotensive rats. Thus, PDE1 appears to be an attractive therapeutic target for the treatment of resistant hypertension, while tachycardia needs to be addressed by further compound structural optimization.
Collapse
Affiliation(s)
- Asim B Dey
- Eli Lilly and Company, Indianapolis, IN, United States
| | - Sherif Khedr
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - James Bean
- Eli Lilly and Company, Indianapolis, IN, United States
| | - Leah L Porras
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | | | - Joseph V Hass
- Eli Lilly and Company, Indianapolis, IN, United States
| | - Xin Zhou
- Eli Lilly and Company, Indianapolis, IN, United States
| | - Maia Terashvili
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Kevin M Ruley
- Eli Lilly and Company, Indianapolis, IN, United States
| | | | - Mark Kowala
- Eli Lilly and Company, Indianapolis, IN, United States
| | - Simon J Atkinson
- Department of Biology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Clement J. Zablocki VA Medical Center, Milwaukee, WI, United States
| | | |
Collapse
|
26
|
Fan L, Gao W, Nguyen BV, Jefferson JR, Liu Y, Fan F, Roman RJ. Impaired renal hemodynamics and glomerular hyperfiltration contribute to hypertension-induced renal injury. Am J Physiol Renal Physiol 2020; 319:F624-F635. [PMID: 32830539 DOI: 10.1152/ajprenal.00239.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Recently, we reported a mutation in γ-adducin (ADD3) was associated with an impaired myogenic response of the afferent arteriole and hypertension-induced chronic kidney disease (CKD) in fawn hooded hypertensive (FHH) rats. However, the mechanisms by which altered renal blood flow (RBF) autoregulation promotes hypertension-induced renal injury remain to be determined. The present study compared the time course of changes in renal hemodynamics and the progression of CKD during the development of DOCA-salt hypertension in FHH 1BN congenic rats [wild-type (WT)] with an intact myogenic response versus FHH 1BN Add3KO (Add3KO) rats, which have impaired myogenic response. RBF was well autoregulated in WT rats but not in Add3KO rats. Glomerular capillary pressure rose by 6 versus 14 mmHg in WT versus Add3KO rats when blood pressure increased from 100 to 150 mmHg. After 1 wk of hypertension, glomerular filtration rate increased by 38% and glomerular nephrin expression decreased by 20% in Add3KO rats. Neither were altered in WT rats. Proteinuria doubled in WT rats versus a sixfold increase in Add3KO rats. The degree of renal injury was greater in Add3KO than WT rats after 3 wk of hypertension. RBF, glomerular filtration rate, and glomerular capillary pressure were lower by 20%, 28%, and 19% in Add3KO rats than in WT rats, which was associated with glomerular matrix expansion and loss of capillary filtration area. The results indicated that impaired RBF autoregulation and eutrophic remodeling of preglomerular arterioles increase the transmission of pressure to glomeruli, which induces podocyte loss and accelerates the progression of CKD in hypertensive Add3KO rats.
Collapse
Affiliation(s)
- Letao Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Wenjun Gao
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bond V Nguyen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Joshua R Jefferson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Yedan Liu
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Richard J Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
27
|
Dasinger JH, Abais-Battad JM, Mattson DL. Influences of environmental factors during preeclampsia. Am J Physiol Regul Integr Comp Physiol 2020; 319:R26-R32. [PMID: 32432917 DOI: 10.1152/ajpregu.00020.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Preeclampsia is a pregnancy-specific disorder that impacts 5-8% of pregnancies and has long-term cardiovascular and metabolic implications for both mother and fetus. The mechanisms are unclear; however, it is believed that preeclampsia is characterized by abnormal vascularization during placentation resulting in the manifestation of clinical signs such as hypertension, proteinuria, and endothelial dysfunction. Although there is no current cure to alleviate the clinical signs, an emerging area of interest in the field is the influence of environmental factors including diet on the risk of preeclampsia. Because preeclampsia has serious cardiovascular implications to both the mother and fetus and most antihypertensive medications are contraindicated in pregnancy, it is important to investigate other potential therapeutic options such as dietary manipulation. The emerging field of nutrigenomics links diet with the gene expression of known pathways such as oxidative stress and inflammation via microbiome-mediated metabolites and could serve as one potential avenue of therapeutic targets for preeclampsia. Although the exact role of nutrition in the pathogenesis of preeclampsia is unknown, this review will focus on known pathways involved in the development of preeclampsia and how dietary intake modulates the microbiome, oxidative stress, and inflammation with an emphasis on nutrigenomics as a potential avenue of further investigation to better understand this pathology.
Collapse
Affiliation(s)
- John Henry Dasinger
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - David L Mattson
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
28
|
Poudel B, Shields CA, Brown AK, Ekperikpe U, Johnson T, Cornelius DC, Williams JM. Depletion of macrophages slows the early progression of renal injury in obese Dahl salt-sensitive leptin receptor mutant rats. Am J Physiol Renal Physiol 2020; 318:F1489-F1499. [PMID: 32390513 DOI: 10.1152/ajprenal.00100.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recently, we reported that obese Dahl salt-sensitive (SS) leptin receptor mutant (SSLepRmutant) rats display progressive renal injury. The present study demonstrated that the early development of renal injury in the SSLepRmutant strain is associated with an increase in the renal infiltration of macrophages compared with lean SS rats. We also examined whether depletion of macrophages with clodronate would reduce the early progression of renal injury in the SSLepRmutant strain. Four-week-old SS and SSLepRmutant rats were treated with either vehicle (PBS) or clodronate (50 mg/kg ip, 2 times/wk) for 4 wk. While the administration of clodronate did not reduce renal macrophage infiltration in SS rats, clodronate decreased macrophages in the kidneys of SSLepRmutant rats by >50%. Interestingly, clodronate significantly reduced plasma glucose, insulin, and triglyceride levels and markedly improved glucose tolerance in SSLepRmutant rats. Treatment with clodronate had no effect on the progression of proteinuria or renal histopathology in SS rats. In the SSLepRmutant strain, proteinuria was markedly reduced during the first 2 wk of treatment (159 ± 32 vs. 303 ± 52 mg/day, respectively). However, after 4 wk of treatment, the effect of clodronate was no longer observed in the SSLepRmutant strain (346 ± 195 vs. 399 ± 50 mg/day, respectively). The kidneys from SSLepRmutant rats displayed glomerular injury with increased mesangial expansion and renal fibrosis versus SS rats. Treatment with clodronate significantly decreased glomerular injury and renal fibrosis in the SSLepRmutant strain. Overall, these data indicate that the depletion of macrophages improves metabolic disease and slows the early progression of renal injury in SSLepRmutant rats.
Collapse
Affiliation(s)
- Bibek Poudel
- Department of Experimental Therapeutics and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Corbin A Shields
- Department of Experimental Therapeutics and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Andrea K Brown
- Department of Experimental Therapeutics and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ubong Ekperikpe
- Department of Experimental Therapeutics and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Tyler Johnson
- Department of Experimental Therapeutics and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Experimental Therapeutics and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan M Williams
- Department of Experimental Therapeutics and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
29
|
Fehrenbach DJ, Dasinger JH, Lund H, Zemaj J, Mattson DL. Splenocyte transfer exacerbates salt-sensitive hypertension in rats. Exp Physiol 2020; 105:864-875. [PMID: 32034948 DOI: 10.1113/ep088340] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/03/2020] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? Recruitment of immune cells to the kidney potentiates hypertensive pathology, but more refined methods are needed to assess these cells functionally. Adoptive transfer studies of immune cells have been limited in rat models and especially in the study of salt-sensitive hypertension. We tested the hypothesis that splenocyte transfer into T-cell-deficient rats is sufficient to exacerbate salt-sensitive hypertension. What is the main finding and its importance? We demonstrate that transfer of splenocytes into T-cell-deficient animals exacerbates salt-sensitive hypertension, and an enrichment in the CD4+ compartment specifically induces this phenomenon. ABSTRACT Increasing evidence of immune system activation during the progression of hypertension and renal injury has led to a need for new methods to study individual cell types. Transfer of immune cells serves as a powerful tool to isolate effects of specific subsets. Transfer studies in Rag1-/- mice have demonstrated an important role of T-cell activation in hypertension, but this approach has yielded limited success in rat models. Using the T-cell-deficient Dahl salt-sensitive (SS) rat, SSCD247-/- , we hypothesized that splenocyte transfer from SS wild-type animals into SSCD247-/- animals would populate the T-cell compartment. The Dahl SS background provides a model for studying salt-sensitive hypertension; therefore, we also tested whether the dietary salt content of the donor would confer differential salt sensitivity in the recipient. To test this, donors were maintained on either a low-salt or a high-salt diet, and at postnatal day 5 the recipients received splenocyte transfer from one of these groups before a high-salt diet challenge. We showed that splenocyte transfer elevated blood pressures while rats were fed low salt and exacerbated the salt-sensitive increase in pressure when they were fed fed high salt. Furthermore, transfer of splenocytes conferred exacerbated renal damage. Lastly, we confirmed the presence of T cells in the circulation and in the spleen, and that infiltration of immune cells, including T cells, macrophages and B cells, into the kidney was elevated in those receiving the transfer. Interestingly, the source of the splenocytes, from donors fed either a low-salt or a high-salt diet, did not significantly affect these salt-sensitive phenotypes.
Collapse
Affiliation(s)
| | - John Henry Dasinger
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, GA, USA
| | - Hayley Lund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeylan Zemaj
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, GA, USA
| |
Collapse
|
30
|
Alsheikh AJ, Dasinger JH, Abais-Battad JM, Fehrenbach DJ, Yang C, Cowley AW, Mattson DL. CCL2 mediates early renal leukocyte infiltration during salt-sensitive hypertension. Am J Physiol Renal Physiol 2020; 318:F982-F993. [PMID: 32150444 DOI: 10.1152/ajprenal.00521.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Studies examining mechanisms of Dahl salt-sensitive (SS) hypertension have implicated the infiltration of leukocytes in the kidneys, which contribute to renal disease and elevated blood pressure. However, the signaling pathways by which leukocytes traffic to the kidneys remain poorly understood. The present study nominated a signaling pathway by analyzing a kidney RNA sequencing data set from SS rats fed either a low-salt (0.4% NaCl) diet or a high-salt (4.0% NaCl) diet. From this analysis, chemokine (C-C motif) ligand 2 (CCL2) and chemokine (C-C motif) receptor 2 (CCR2) were nominated as a potential pathway modifying renal leukocyte infiltration and contributing to SS hypertension. The functional role of the CCL2/CCR2 pathway was tested by daily administration of CCR2 antagonist (RS-102895 at 5 mg·kg-1·day-1 in DMSO) or DMSO vehicle for 3 or 21 days by intraperitoneal injections during the high salt challenge. Blood pressure, renal leukocyte infiltration, and renal damage were evaluated. The results demonstrated that RS-102895 treatment ameliorated renal damage (urinary albumin excretion; 43.4 ± 5.1 vs. 114.7 ± 15.2 mg/day in vehicle, P < 0.001) and hypertension (144.3 ± 2.2 vs. 158.9 ± 4.8 mmHg in vehicle, P < 0.001) after 21 days of high-salt diet. It was determined that renal leukocyte infiltration was blunted by day 3 of the high-salt diet (1.4 ± 0.1 vs. 1.9 ± 0.2 in vehicle × 106 CD45+ cells/kidney, P = 0.034). An in vitro chemotaxis assay validated the effect of RS-102895 on leukocyte chemotaxis toward CCL2. The results suggest that increased CCL2 in SS kidneys is important in the early recruitment of leukocytes, and blockade of this recruitment by administering RS-102895 subsequently blunted the renal damage and hypertension.
Collapse
Affiliation(s)
- Ammar J Alsheikh
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - John Henry Dasinger
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Justine M Abais-Battad
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Daniel J Fehrenbach
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
31
|
McPherson KC, Shields CA, Poudel B, Johnson AC, Taylor L, Stubbs C, Nichols A, Cornelius DC, Garrett MR, Williams JM. Altered renal hemodynamics is associated with glomerular lipid accumulation in obese Dahl salt-sensitive leptin receptor mutant rats. Am J Physiol Renal Physiol 2020; 318:F911-F921. [PMID: 32068459 DOI: 10.1152/ajprenal.00438.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The present study examined whether development of renal injury in the nondiabetic obese Dahl salt-sensitive leptin receptor mutant (SSLepRmutant) strain is associated with elevations in glomerular filtration rate and renal lipid accumulation. Baseline mean arterial pressure at 6 wk of age was similar between Dahl salt-sensitive wild-type (SSWT) and SSLepRmutant rats. However, by 18 wk of age, the SSLepRmutant strain developed hypertension, while the elevation in mean arterial pressure was not as severe in SSWT rats (192 ± 4 and 149 ± 6 mmHg, respectively). At baseline, proteinuria was fourfold higher in SSLepRmutant than SSWT rats and remained elevated throughout the study. The early development of progressive proteinuria was associated with renal hyperfiltration followed by a decline in renal function over the course of study in the SSLepRmutant compared with SSWT rats. Kidneys from the SSLepRmutant strain displayed more glomerulosclerosis and glomerular lipid accumulation than SSWT rats. Glomeruli were isolated from the renal cortex of both strains at 6 and 18 wk of age, and RNA sequencing was performed to identify genes and pathways driving glomerular injury. We observed significant increases in expression of the influx lipid transporters, chemokine (C-X-C motif) ligand 16 (Cxcl16) and scavenger receptor and fatty acid translocase (Cd36), respectively, and a significant decrease in expression of the efflux lipid transporter, ATP-binding cassette subfamily A member 2 (Abca2; cholesterol efflux regulatory protein 2), in SSLepRmutant compared with SSWT rats at 6 and 18 wk of age, which were validated by RT-PCR analysis. These data suggest an association between glomerular hyperfiltration and glomerular lipid accumulation during the early development of proteinuria associated with obesity.
Collapse
Affiliation(s)
- Kasi C McPherson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Corbin A Shields
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bibek Poudel
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Ashley C Johnson
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Lateia Taylor
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Cassandra Stubbs
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Alyssa Nichols
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Denise C Cornelius
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Emergency Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael R Garrett
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jan M Williams
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
32
|
Fehrenbach DJ, Mattson DL. Inflammatory macrophages in the kidney contribute to salt-sensitive hypertension. Am J Physiol Renal Physiol 2020; 318:F544-F548. [PMID: 31984790 DOI: 10.1152/ajprenal.00454.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This review will highlight recent studies that have investigated the relationship between Na+, renal macrophage polarization, and renal damage. A hyperosmotic environment drives the macrophage toward a proinflammatory phenotype and away from an anti-inflammatory phenotype. Animal models of salt-sensitive hypertension demonstrate a characteristic infiltration of macrophages into the kidney that is greatly reduced when blood pressure is lowered. Because general immunosuppression or macrophage depletion leads to a host of adverse side effects, more recent studies have modulated the interaction of specific signaling molecules, including NOD-like receptor family pyrin domain-containing 3, chemokine (C-X-C motif) ligand 16, and VEGF, to prevent the end-organ renal damage that accumulates in salt-sensitive disease.
Collapse
Affiliation(s)
- Daniel J Fehrenbach
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, Georgia
| | - David L Mattson
- Department of Physiology, Augusta University and the Medical College of Georgia, Augusta, Georgia
| |
Collapse
|
33
|
Abais-Battad JM, Lund H, Dasinger JH, Fehrenbach DJ, Cowley AW, Mattson DL. NOX2-derived reactive oxygen species in immune cells exacerbates salt-sensitive hypertension. Free Radic Biol Med 2020; 146:333-339. [PMID: 31730933 PMCID: PMC6942201 DOI: 10.1016/j.freeradbiomed.2019.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/29/2019] [Accepted: 11/11/2019] [Indexed: 12/11/2022]
Abstract
Previous studies utilizing the SSp67phox-/- rat have demonstrated the importance of systemic NADPH oxidase NOX2-derived reactive oxygen species (ROS) production in the pathogenesis of Dahl Salt-Sensitive (SS) hypertension and renal damage. It is established that the immune system contributes to the development of SS hypertension and our laboratory has observed an enrichment of NOX2 subunits in infiltrating T cells. However, the contribution of immune cell-derived ROS in SS hypertension remains unknown. To assess the role of ROS in immune cells, SSp67phox-/- rats underwent total body irradiation and received bone marrow transfer from either SS (+SS) or SSp67phox-/- (+SSp67phox-/-) donor rats. Demonstrated in a respiratory burst assay, response to phorbol 12-myristate 13-acetate stimulus (135 μM) was 10.2-fold greater in peritoneal macrophages isolated from +SS rats compared to nonresponsive + SSp67phox-/- cells, validating that + SS rats were capable of producing NOX2-derived ROS in cells of hematopoietic origin. After 3 weeks of high salt challenge, there was an exacerbated increase in mean arterial pressure in +SS rats compared to + SSp67phox-/- control rats (176.1 ± 4.7 vs 147.9 ± 8.4 mmHg, respectively), which was accompanied by a significant increase in albuminuria (168.3 ± 23.7 vs 107.0 ± 20.4 mg/day) and renal medullary protein cast formation (33.2 ± 4.7 vs 8.1 ± 3.5%). Interestingly, upon analysis of renal immune cells, there was trending increase of CD11b/c + monocytes and macrophages in the kidney of +SS rats (4.7 ± 0.4 vs 3.5 ± 0.5 × 106 cells/kidney, +SS vs + SSp67phox-/-, p = 0.06). These data altogether demonstrate that immune cell production of NOX2-derived ROS is sufficient to exacerbate Dahl SS hypertension, renal damage, and renal inflammation.
Collapse
Affiliation(s)
- Justine M Abais-Battad
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Hayley Lund
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - John Henry Dasinger
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Daniel J Fehrenbach
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Allen W Cowley
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
34
|
Dasinger JH, Alsheikh AJ, Abais-Battad JM, Pan X, Fehrenbach DJ, Lund H, Roberts ML, Cowley AW, Kidambi S, Kotchen TA, Liu P, Liang M, Mattson DL. Epigenetic Modifications in T Cells: The Role of DNA Methylation in Salt-Sensitive Hypertension. Hypertension 2019; 75:372-382. [PMID: 31838911 DOI: 10.1161/hypertensionaha.119.13716] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The SS (Dahl salt sensitive) rat is an established model of hypertension and renal damage that is accompanied with immune system activation in response to a high-salt diet. Investigations into the effects of sodium-independent and dependent components of the diet were shown to affect the disease phenotype with SS/MCW (JrHsdMcwi) rats maintained on a purified diet (AIN-76A) presenting with a more severe phenotype relative to grain-fed SS/CRL (JrHsdMcwiCrl) rats. Since contributions of the immune system, environment, and diet are documented to alter this phenotype, this present study examined the epigenetic profile of T cells isolated from the periphery and the kidney from these colonies. T cells isolated from kidneys of the 2 colonies revealed that transcriptomic and functional differences may contribute to the susceptibility of hypertension and renal damage. In response to high-salt challenge, the methylome of T cells isolated from the kidney of SS/MCW exhibit a significant increase in differentially methylated regions with a preference for hypermethylation compared with the SS/CRL kidney T cells. Circulating T cells exhibited similar methylation profiles between colonies. Utilizing transcriptomic data from T cells isolated from the same animals upon which the DNA methylation analysis was performed, a predominant negative correlation was observed between gene expression and DNA methylation in all groups. Lastly, inhibition of DNA methyltransferases blunted salt-induced hypertension and renal damage in the SS/MCW rats providing a functional role for methylation. This study demonstrated the influence of epigenetic modifications to immune cell function, highlighting the need for further investigations.
Collapse
Affiliation(s)
- John Henry Dasinger
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Ammar J Alsheikh
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Justine M Abais-Battad
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Xiaoqing Pan
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Daniel J Fehrenbach
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Hayley Lund
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Michelle L Roberts
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Allen W Cowley
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Srividya Kidambi
- Medicine (S.K., T.A.K.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Theodore A Kotchen
- Medicine (S.K., T.A.K.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Pengyuan Liu
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - Mingyu Liang
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| | - David L Mattson
- From the Departments of Physiology (J.H.D., A.J.A., J.M.A.-B., X.P., D.J.F., H.L., M.L.R., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin Milwaukee, Wisconsin, USA
| |
Collapse
|