1
|
Johnson K, Bray JF, Heaps CL. Sexually dimorphic mechanisms of H 2O 2-mediated dilation in porcine coronary arterioles with ischemia and endurance exercise training. J Appl Physiol (1985) 2025; 138:950-963. [PMID: 40059640 DOI: 10.1152/japplphysiol.00761.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/28/2024] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
We determined the impact of sex on H2O2-mediated dilation in coronary arterioles and the contribution of K+ channels after exercise training in ischemic heart disease. We hypothesized that arterioles from male and female swine would similarly display impaired H2O2-induced dilation after chronic occlusion that would be corrected by exercise training. Yucatan miniswine were surgically instrumented with an ameroid constrictor around the proximal left circumflex artery, gradually inducing occlusion and a collateral-dependent myocardium. Arterioles from the left anterior descending artery myocardial region served as nonoccluded controls. Eight weeks postoperatively, swine of each sex were separated into sedentary and exercise-trained (progressive treadmill regimen; 5 days/wk for 14 wk) groups. Collateral-dependent arterioles of sedentary female pigs displayed impaired sensitivity to H2O2 that was reversed with exercise training. In contrast, male pigs exhibited enhanced sensitivity to H2O2 in collateral-dependent versus nonoccluded arterioles in both sedentary and exercise-trained groups. Large-conductance, calcium-dependent K+ (BKCa) and 4-aminopyridine (AP)-sensitive voltage-gated K+ (Kv) channels contributed to H2O2-mediated dilation in nonoccluded and collateral-dependent arterioles of exercise-trained females, but not in arterioles of sedentary female or sedentary or exercise-trained male swine. BKCa channel, protein kinase A (PKA), and protein kinase G (PKG) protein levels were not significantly different between groups, nor were kinase enzymatic activities. Taken together, our studies suggest that in female swine, exercise training stimulates the coupling of H2O2 signaling with BKCa and 4-AP-sensitive Kv channels, compensating for impaired dilation in collateral-dependent arterioles. Interestingly, coronary arterioles from neither sedentary female or male swine, regardless of training status, depended upon BKCa or 4-AP-sensitive Kv channels for H2O2-mediated dilation.NEW & NOTEWORTHY The current studies reveal sexually dimorphic adaptations to H2O2-mediated dilation, and unique contributions of K+ channels, in coronary arterioles from swine subjected to chronic ischemia and exercise training; findings important for development of therapeutic strategies. In female swine, chronic ischemia attenuates dilation, which is reversed by exercise training via BKCa and Kv channel stimulation. In male swine, ischemia enhances dilation to H2O2, which is further augmented by exercise training and independent of BKCa and Kv channels.
Collapse
Affiliation(s)
- Kalen Johnson
- Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Jeff F Bray
- Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| | - Cristine L Heaps
- Department of Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
- Michael E. DeBakey Institute for Comparative Cardiovascular Science and Biomedical Devices, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States
| |
Collapse
|
2
|
Martínez-García A, Pérez JA, Tapia-Castillo A, Hernández MP, Solórzano M, Carrión P, Fardella CE, Carvajal CA. Influence of estradiol deficiency on the mineralocorticoid receptor response in postmenopausal women: a cross-sectional study. Climacteric 2025; 28:162-168. [PMID: 39928302 DOI: 10.1080/13697137.2025.2455181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVE Premenopausal women (PreM) have a cardioprotective advantage over postmenopausal women (PostM) due to estrogen. The interaction of estrogen with the mineralocorticoid receptor (MR) pathway remains unexplored. This study aimed to identify changes in aldosterone, renin and sexual steroid levels and MR surrogate biomarkers in PostM that may explain changes in blood pressure and renal damage. METHODS A cross-sectional study was carried out with 47 normotensive and hypertensive Chilean women distributed between PreM and PostM. Clinical, anthropometric and biochemical parameters, including aldosterone, plasma renin activity (PRA) and surrogate markers of MR activity, were assessed. RESULTS PostM had greater systolic blood pressure (SBP) (p < 0.001) than PreM. A negative correlation was observed between estradiol and fractional excretion of potassium (FEK) (ρ = -0.29; p = 0.023), adjusted for age and SBP. Compared with hypertensive PreM, hypertensive PostM (PostM-HT) showed reduced PRA (p = 0.045) and greater FEK (p = 0.04). Normotensive PostM (Post-NT) exhibited greater SBP (p = 0.03), neutrophil gelatinase-associated lipocalin (NGAL) levels (p = 0.04) and FEK (p = 0.03) than normotensive PreM. CONCLUSION Our results suggest enhanced MR sensitivity not only in PostM-HT, as evidenced by lower PRA and elevated FEK, but also in PostM-NT, who exhibited greater FEK and NGAL levels, surrogate markers of MR activation. These results support a novel role of MR activation and cardiovascular risk in PostM women.
Collapse
Affiliation(s)
- Alejandra Martínez-García
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Pérez
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Tapia-Castillo
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María P Hernández
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marlin Solórzano
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Carrión
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos E Fardella
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian A Carvajal
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Translational Center for Endocrinology (CETREN), Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
3
|
Turner CG, Stanhewicz AE, Nielsen KE, Otis JS, Feresin RG, Wong BJ. Oral contraceptive pill phase alters mechanisms contributing to cutaneous microvascular function in response to local heating. Am J Physiol Regul Integr Comp Physiol 2025; 328:R374-R385. [PMID: 39938890 DOI: 10.1152/ajpregu.00159.2024] [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: 06/13/2024] [Revised: 07/16/2024] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The purpose of this study was to investigate the effect of oral contraceptive pill (OCP) phase on in vivo microvascular endothelium-dependent vasodilation and contributions of nitric oxide (NO), cyclooxygenase (COX), and endothelial-derived hyperpolarizing factors (EDHFs). Participants completed two experimental visits in random order, during the 1) low and 2) high hormone phase of the OCP cycle. Endothelium-dependent dilation was assessed in the cutaneous microvasculature via local heating at four intradermal microdialysis sites treated with: 1) lactated Ringer's (control), 2) 10 mM ketorolac (Keto, COX inhibitor), 3) 50 mM tetraethylammonium (TEA, calcium-activated potassium channel inhibitor), and 4) 10 mM ketorolac + 50 mM TEA (Keto + TEA). Perfusion of 20 mM Nω-nitro-l-arginine methyl ester (l-NAME) at each site was used to quantify the l-NAME-sensitive component of dilation, suggesting NO contribution. There was no effect of OCP phase on endothelium-dependent dilation (P = 0.75) or the l-NAME-sensitive component of the response (P = 0.09, d = 0.7) at control sites. Inhibition of COX increased baseline blood flow regardless of OCP phase (all P < 0.01). Control and Keto sites elicited greater endothelium-dependent dilation than TEA and Keto + TEA sites in both phases (all P < 0.0001). During the low hormone phase, the l-NAME-sensitive component was greater at control compared with TEA sites (P < 0.01). During the high hormone phase, the l-NAME-sensitive component was greater at Keto compared with TEA sites (P < 0.01). Within-participant differences between control and Keto sites support a phase-dependent restraint of NO activity via COX pathways (P = 0.01). These findings demonstrate that the OCP phase affects underlying mechanistic pathways contributing to cutaneous microvascular endothelial function.NEW & NOTEWORTHY This study investigates the effect of OCP phase on in vivo microvascular endothelium-dependent vasodilation and explores underlying mechanisms. Present findings suggest OCP phase does not affect overall microvascular endothelium-dependent dilation but does affect the underlying mechanisms. In women using OCP, there is a robust reliance on EDHF pathways and the COX pathway moderates basal microvascular blood flow and demonstrates a phase-dependent restraint of the NO pathway.
Collapse
Affiliation(s)
- Casey G Turner
- Department of Kinesiology and Health, Georgia State University, Atlanta, Georgia, United States
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States
| | - Anna E Stanhewicz
- Department of Health and Human Physiology, University of Iowa, Iowa City, Iowa, United States
| | - Karen E Nielsen
- Department of Population Health Sciences, School of Public Health, Georgia State University, Atlanta, Georgia, United States
| | - Jeffrey S Otis
- Department of Kinesiology and Health, Georgia State University, Atlanta, Georgia, United States
| | - Rafaela G Feresin
- Department of Nutrition, Georgia State University, Atlanta, Georgia, United States
| | - Brett J Wong
- Department of Kinesiology and Health, Georgia State University, Atlanta, Georgia, United States
| |
Collapse
|
4
|
Camarda ND, Lu Q, Tesfu AF, Liu RR, Ibarrola J, Jaffe IZ. Mineralocorticoid Receptor in Endothelial Cells Contributes to Vascular Endothelial Growth Factor Receptor Inhibitor-Induced Vascular and Kidney Damage. Am J Hypertens 2025; 38:104-110. [PMID: 39514632 PMCID: PMC11735467 DOI: 10.1093/ajh/hpae140] [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: 06/13/2024] [Revised: 08/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Vascular endothelial growth factor receptor inhibitors (VEGFRis) improve cancer patient survival by inhibiting tumor angiogenesis. However, VEGFRis induce treatment-limiting hypertension which has been associated with impaired vascular endothelial cell (EC) function and kidney damage. The mineralocorticoid receptor (MR) regulates blood pressure (BP) via its effects on the vasculature and the kidney. Thus, we interrogated the role of the MR in EC dysfunction, renal impairment, and hypertension in a mouse model of VEGFRi-induced hypertension using sorafenib. METHODS EC dysfunction in mesenteric arterioles was assessed by immunoblotting for phosphorylation of endothelial nitric oxide synthase (eNOS) at serine 1177. Renal damage was measured by assessing glomerular endotheliosis histologically. BP was measured using implanted radiotelemetry. RESULTS Six days of sorafenib treatment significantly impaired mesenteric resistance vessel EC function, induced renal damage, and increased BP. Pharmacologic MR blockade with spironolactone prevented the sorafenib-induced decline in eNOS phosphorylation and renal glomerular endotheliosis, without affecting systolic BP (SBP) or diastolic BP. Mice with the MR knocked out specifically in ECs (EC-MR-KO) were protected from sorafenib-induced EC dysfunction and glomerular endotheliosis, whereas smooth muscle cell-specific MR (SMC-MR) knockout mice were not. Neither EC-MR nor SMC-MR knockout affected the degree to which sorafenib increased SBP or diastolic BP. CONCLUSIONS These results reveal that the MR, specifically in EC but not in SMCs, is necessary for VEGFRi-induced renal and vascular injury. While ineffective at lowering SBP, these data suggest potential therapeutic benefits of MR antagonists, like spironolactone, to protect the vasculature and the kidneys from VEGFRi-induced injury.
Collapse
Affiliation(s)
- Nicholas D Camarda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
- Genetics, Molecular, and Cellular Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Angelina F Tesfu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Rui R Liu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
- Genetics, Molecular, and Cellular Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Liu J, Zheng Z, Sun J, Gu X, Yu X, Wang Y, Yu X. Conjunctival microvascular alteration in patients with coronary artery disease assessed using optical coherence tomographic angiography. Microvasc Res 2025; 157:104733. [PMID: 39236912 DOI: 10.1016/j.mvr.2024.104733] [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: 05/28/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND To quantify conjunctival microvascular characteristics obtained by optical coherence tomographic angiography (OCTA) and investigate their relationship with the presence and severity of coronary artery disease (CAD). METHODS This cross-sectional study included 103 consecutive CAD patients confirmed by coronary angiography and 125 non-CAD controls. The temporal conjunctivas along the limbus of each participant were scanned using OCTA. Quantification of conjunctival microvasculature was performed by AngioTool software. The severity of the disease was evaluated using SYNTAX and Gensini scores. RESULTS Compared to the controls, the CAD group exhibited significantly lower vessel area density (30.22 ± 3.34 vs. 26.70 ± 4.43 %, p < 0.001), lower vessel length density (6.39 ± 0.77 vs. 5.71 ± 0.89/m, p < 0.001), lower junction density (3.44 ± 0.56 vs. 3.05 ± 0.63/m, p < 0.001), and higher lacunarity (0.11 ± 0.03 vs. 0.14 ± 0.05, p < 0.001). Among all participants, lower vessel area density, lower vessel length density, lower junction density, and higher lacunarity were associated with greater odds of having CAD; the adjusted ORs (95 % confidence intervals) per one SD decrease were 2.71 (1.71, 4.29), 2.51(1.61, 3.90), 2.06 (1.39, 3.05), and 0.36 (0.23, 0.58), respectively. Among CAD patients, junction density was negatively associated with the Gensini score (r = -0.359, p = 0.037) and the Syntax score (r = -0.350, p = 0.042) in women but not in men (p > 0.05). CONCLUSIONS Conjunctival microvascular characteristics were significantly associated with the presence of CAD. Junction density significantly associated with the severity of CAD among women patients.
Collapse
Affiliation(s)
- Jing Liu
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhaoxia Zheng
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiayi Sun
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoya Gu
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xue Yu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanling Wang
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Xiaobing Yu
- Department of Ophthalmology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Santos J, La Fuente JM, Fernández A, Ruano P, Angulo J. LDL-c/HDL-c Ratio and NADPH-Oxidase-2-Derived Oxidative Stress as Main Determinants of Microvascular Endothelial Function in Morbidly Obese Subjects. Antioxidants (Basel) 2024; 13:1139. [PMID: 39334798 PMCID: PMC11444145 DOI: 10.3390/antiox13091139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
The identification of obese subjects at higher risk for cardiovascular disease (CVD) is required. We aimed to characterize determinants of endothelial dysfunction, the initial step to CVD, in small omental arteries of visceral fat from obese subjects. The influences of analytical parameters and vascular oxidative stress mediated by NADPH-oxidase-2 (NOX2) on endothelial function were determined. Specimens were obtained from 51 obese subjects undergoing bariatric surgery and 14 non-obese subjects undergoing abdominal surgery. Obese subjects displayed reduced endothelial vasodilation to bradykinin (BK). Endothelial vasodilation (pEC50 for BK) among obese subjects was significantly and negatively associated with low-density lipoprotein cholesterol (LDL-c)/high-density lipoprotein cholesterol (HDL-c) ratio (r = -0.510, p = 0.0001) in both women and men, while other metabolic parameters and comorbidities failed to predict endothelial function. The vascular expression of NOX2 was upregulated in obese subjects and was related to decreased endothelial vasodilation (r = -0.529, p = 0.0006, n = 38) and increased oxidative stress (r = 0.783, p = 0.0044, n = 11) in arterial segments. High LDL-c/HDL-c (>2) and high NOX2 (above median) were independently associated with reduced endothelial function, but the presence of both conditions was related to a further impairment. Concomitant elevated LDL-c/HDL-c ratio and high vascular expression of NOX2 would exacerbate endothelial impairment in obesity and could reveal a deleterious profile for cardiovascular outcomes among obese subjects.
Collapse
Affiliation(s)
- Jorge Santos
- Unidade de Cirurgia Esofagogástrica e Tratamento Cirúrgico de Obesidade, Centro Hospitalar e Universitário de Santo António (CHUdSA), 4099-001 Porto, Portugal
| | - José M La Fuente
- Serviço de Urologia, Centro Hospitalar e Universitário de Santo António (CHUdSA), 4099-001 Porto, Portugal
| | - Argentina Fernández
- Servicio de Histología-Investigación. Unidad de Investigación Traslacional en Cardiología-IRYCIS/UFV, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Paula Ruano
- Servicio de Histología-Investigación. Unidad de Investigación Traslacional en Cardiología-IRYCIS/UFV, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Javier Angulo
- Servicio de Histología-Investigación. Unidad de Investigación Traslacional en Cardiología-IRYCIS/UFV, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
7
|
Vecchiola A, Uslar T, Friedrich I, Aguirre J, Sandoval A, Carvajal CA, Tapia-Castillo A, Martínez-García A, Fardella CE. The role of sex hormones in aldosterone biosynthesis and their potential impact on its mineralocorticoid receptor. Cardiovasc Endocrinol Metab 2024; 13:e0305. [PMID: 38846628 PMCID: PMC11155591 DOI: 10.1097/xce.0000000000000305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/22/2024] [Indexed: 06/09/2024]
Abstract
Blood pressure (BP) regulation is a complex process involving various hormones, including aldosterone and its mineralocorticoid receptor. Mineralocorticoid receptor is expressed in several tissues, including the kidney, and plays a crucial role in regulating BP by controlling the sodium and water balance. During different stages of life, hormonal changes can affect mineralocorticoid receptor activity and aldosterone levels, leading to changes in BP. Increasing evidence suggests that sex steroids modulate aldosterone levels. Estrogens, particularly estradiol, mediate aldosterone biosynthesis by activating classical estrogen receptors and the G protein-coupled receptor. Progesterone acts as an anti-mineralocorticoid by inhibiting the binding of aldosterone to the mineralocorticoid receptor. Moreover, progesterone inhibits aldosterone synthase enzymes. The effect of testosterone on aldosterone synthesis is still a subject of debate. However, certain studies show that testosterone downregulates the mRNA levels of aldosterone synthase, leading to decreased plasma aldosterone levels.
Collapse
Affiliation(s)
- Andrea Vecchiola
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Thomas Uslar
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Isidora Friedrich
- Departamento de Endocrinologìa, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago
| | - Joaquin Aguirre
- Departamento de Endocrinologìa, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago
| | - Alejandra Sandoval
- Escuela de Tecnología Médica, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Cristian A. Carvajal
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Alejandra Tapia-Castillo
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Alejandra Martínez-García
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| | - Carlos E. Fardella
- Departamento de Endocrinología, Facultad de Medicina, Centro Traslacional de Endocrinología UC (CETREN), Pontificia Universidad Católica de Chile
| |
Collapse
|
8
|
Moronge D, Ayulo V, Elgazzaz M, Mellott E, Ogbi S, Faulkner JL. Both endothelial mineralocorticoid receptor expression and hyperleptinemia are required for clinical characteristics of placental ischemia in mice. Am J Physiol Heart Circ Physiol 2024; 327:H118-H130. [PMID: 38758130 PMCID: PMC11380964 DOI: 10.1152/ajpheart.00188.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/23/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
One of the initiating events in preeclampsia (PE) is placental ischemia. Rodent models of placental ischemia do not present with vascular endothelial dysfunction, a hallmark of PE. We previously demonstrated a role for leptin in endothelial dysfunction in pregnancy in the absence of placental ischemia. We hypothesized that placental ischemia requires hyperleptinemia and endothelial mineralocorticoid receptor (ECMR) expression to induce PE-associated endothelial dysfunction in pregnant mice. We induced placental ischemia via the reduced uterine perfusion pressure (RUPP) procedure in pregnant ECMR-intact (ECMR+/+) and ECMR deletion (ECMR-/-) mice at gestational day (GD) 13. ECMR+/+ RUPP pregnant mice also received concurrent leptin infusion via miniosmotic pump (0.9 mg/kg/day). RUPP increased blood pressure via radiotelemetry and decreased fetal growth in ECMR+/+ pregnant mice. Both increases in blood pressure and reduced fetal growth were abolished in RUPP ECMR-/- mice. Placental ischemia did not decrease endothelial-dependent relaxation to acetylcholine (ACh) but increased phenylephrine (Phe) contraction in mesenteric arteries of pregnant mice, which was ablated by ECMR deletion. Addition of leptin to RUPP mice significantly reduced ACh relaxation in ECMR+/+ pregnant mice, accompanied by an increase in soluble FMS-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PLGF) ratio. In conclusion, our data indicate that high leptin levels drive endothelial dysfunction in PE and that ECMR is required for clinical characteristics of hypertension and fetal growth restriction in placental ischemia PE. Collectively, we show that both ECMR and leptin play a role to mediate PE.NEW & NOTEWORTHY Leptin is a key feature of preeclampsia that initiates vascular endothelial dysfunction in preeclampsia characterized by placental ischemia. Endothelial mineralocorticoid receptor (ECMR) deletion in placental ischemia protects pregnant mice from elevations in blood pressure and fetal growth restriction in pregnancy. Increases in leptin production mediate the key pathological feature of endothelial dysfunction in preeclampsia in rodents. ECMR activation contributes to the increase in blood pressure and fetal growth restriction in preeclampsia.
Collapse
Affiliation(s)
- Desmond Moronge
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Victor Ayulo
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Pediatrics, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Mona Elgazzaz
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Elisabeth Mellott
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Safia Ogbi
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
- Department of Obstetrics and Gynecology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
9
|
Ibarrola J, Jaffe IZ. The Mineralocorticoid Receptor in the Vasculature: Friend or Foe? Annu Rev Physiol 2024; 86:49-70. [PMID: 37788489 DOI: 10.1146/annurev-physiol-042022-015223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Originally described as the renal aldosterone receptor that regulates sodium homeostasis, it is now clear that mineralocorticoid receptors (MRs) are widely expressed, including in vascular endothelial and smooth muscle cells. Ample data demonstrate that endothelial and smooth muscle cell MRs contribute to cardiovascular disease in response to risk factors (aging, obesity, hypertension, atherosclerosis) by inducing vasoconstriction, vascular remodeling, inflammation, and oxidative stress. Extrapolating from its role in disease, evidence supports beneficial roles of vascular MRs in the context of hypotension by promoting inflammation, wound healing, and vasoconstriction to enhance survival from bleeding or sepsis. Advances in understanding how vascular MRs become activated are also reviewed, describing transcriptional, ligand-dependent, and ligand-independent mechanisms. By synthesizing evidence describing how vascular MRs convert cardiovascular risk factors into disease (the vascular MR as a foe), we postulate that the teleological role of the MR is to coordinate responses to hypotension (the MR as a friend).
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA;
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA;
| |
Collapse
|
10
|
Looft-Wilson RC, Stechmann JK, Milenski KG, Shah VM, Kulkarni PG, Arif AB, Guiot T, Beinlich NMC, Dos Santos CA, Rice SK. Myoendothelial feedback in mouse mesenteric resistance arteries is similar between the sexes, dependent on nitric oxide synthase, and independent of TPRV4. Am J Physiol Heart Circ Physiol 2024; 326:H190-H202. [PMID: 37921665 PMCID: PMC11213485 DOI: 10.1152/ajpheart.00170.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 10/17/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
Myoendothelial feedback (MEF), the endothelium-dependent vasodilation following sympathetic vasoconstriction (mediated by smooth muscle to endothelium gap junction communication), has been well studied in resistance arteries of males, but not females. We hypothesized that MEF responses would be similar between the sexes, but different in the relative contribution of the underlying nitric oxide and hyperpolarization mechanisms, given that these mechanisms differ between the sexes in agonist-induced endothelium-dependent dilation. We measured MEF responses (diameter changes) of male and female first- to second-order mouse mesenteric arteries to phenylephrine (10 µM) over 30 min using isolated pressure myography ± blinded inhibition of nitric oxide synthase (NOS) using Nω-nitro-l-arginine methyl ester (l-NAME; 0.1-1.0 mM), hyperpolarization using 35 mM KCl, or transient receptor potential vanilloid 4 (TRPV4) channels using GSK219 (0.1-1.0 µM) or RN-1734 (30 µM). MEF was similar [%dilation (means ± SE): males = 26.7 ± 2.0 and females = 26.1 ± 1.9 at 15 min] and significantly inhibited by l-NAME (1.0 mM) at 15 min [%dilation (means ± SE): males = 8.2 ± 3.3, P < 0.01; females = 6.8 ± 1.9, P < 0.001] and over time (P < 0.01) in both sexes. l-NAME (0.1 mM) + 35 mM KCl nearly eliminated MEF in both sexes (P < 0.001-0.0001). Activation of TRPV4 with GSK101 (0.1-10 µM) induced similar dilation between the sexes. Inhibition of TRPV4, which is reportedly involved in the hyperpolarization mechanism, did not inhibit MEF in either sex. Similar expression of eNOS was found between the sexes with Western blot. Thus, MEF is prominent and similar in murine first- and second-order mesenteric resistance arteries of both sexes, and reliant primarily on NOS and secondarily on hyperpolarization, but not TRPV4.NEW & NOTEWORTHY We found that female mesenteric resistance arteries have similar postconstriction dilatory responses (i.e., myoendothelial feedback) to a sympathetic neurotransmitter analog as male arteries. Both sexes use nitric oxide synthase (NOS) and hyperpolarization, but not TRPV4, in this response. Moreover, the key protein involved in this pathway (eNOS) is similarly expressed in these arteries between the sexes. These similarities are surprising given that agonist-induced endothelium-dependent dilatory mechanisms differ in these arteries between the sexes.
Collapse
Affiliation(s)
- Robin C Looft-Wilson
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| | - Jacob K Stechmann
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| | - Katherine G Milenski
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| | - Vishakha M Shah
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| | - Preetika G Kulkarni
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| | - Arusha B Arif
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| | - Tanner Guiot
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| | | | | | - Spencer K Rice
- Department of Kinesiology, William and Mary, Williamsburg, Virginia, United States
| |
Collapse
|
11
|
Jia G, Hill MA, Sowers JR. Vascular endothelial mineralocorticoid receptors and epithelial sodium channels in metabolic syndrome and related cardiovascular disease. J Mol Endocrinol 2023; 71:e230066. [PMID: 37610001 PMCID: PMC10502958 DOI: 10.1530/jme-23-0066] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023]
Abstract
Metabolic syndrome is a group of risk factors that increase the risk of developing metabolic and cardiovascular disease (CVD) and include obesity, dyslipidemia, insulin resistance, atherosclerosis, hypertension, coronary artery disease, and heart failure. Recent research indicates that excessive production of aldosterone and associated activation of mineralocorticoid receptors (MR) impair insulin metabolic signaling, promote insulin resistance, and increase the risk of developing metabolic syndrome and CVD. Moreover, activation of specific epithelial sodium channels (ENaC) in endothelial cells (EnNaC), which are downstream targets of endothelial-specific MR (ECMR) signaling, are also believed to play a crucial role in the development of metabolic syndrome and CVD. These adverse effects of ECMR/EnNaC activation are mediated by increased oxidative stress, inflammation, and lipid metabolic disorders. It is worth noting that ECMR/EnNaC activation and the pathophysiology underlying metabolic syndrome and CVD appears to exhibit sexual dimorphism. Targeting ECMR/EnNaC signaling may have a beneficial effect in preventing insulin resistance, diabetes, metabolic syndrome, and related CVD. This review aims to examine our current understanding of the relationship between MR activation and increased metabolic syndrome and CVD, with particular emphasis placed on the role for endothelial-specific ECMR/EnNaC signaling in these pathological processes.
Collapse
Affiliation(s)
- Guanghong Jia
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - James R Sowers
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| |
Collapse
|
12
|
Dona MSI, Hsu I, Meuth AI, Brown SM, Bailey CA, Aragonez CG, Russell JJ, Krstevski C, Aroor AR, Chandrasekar B, Martinez-Lemus LA, DeMarco VG, Grisanti LA, Jaffe IZ, Pinto AR, Bender SB. Multi-omic analysis of the cardiac cellulome defines a vascular contribution to cardiac diastolic dysfunction in obese female mice. Basic Res Cardiol 2023; 118:11. [PMID: 36988733 PMCID: PMC10060343 DOI: 10.1007/s00395-023-00983-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/30/2023]
Abstract
Coronary microvascular dysfunction (CMD) is associated with cardiac dysfunction and predictive of cardiac mortality in obesity, especially in females. Clinical data further support that CMD associates with development of heart failure with preserved ejection fraction and that mineralocorticoid receptor (MR) antagonism may be more efficacious in obese female, versus male, HFpEF patients. Accordingly, we examined the impact of smooth muscle cell (SMC)-specific MR deletion on obesity-associated coronary and cardiac diastolic dysfunction in female mice. Obesity was induced in female mice via western diet (WD) feeding alongside littermates fed standard diet. Global MR blockade with spironolactone prevented coronary and cardiac dysfunction in obese females and specific deletion of SMC-MR was sufficient to prevent obesity-associated coronary and cardiac diastolic dysfunction. Cardiac gene expression profiling suggested reduced cardiac inflammation in WD-fed mice with SMC-MR deletion independent of blood pressure, aortic stiffening, and cardiac hypertrophy. Further mechanistic studies utilizing single-cell RNA sequencing of non-cardiomyocyte cell populations revealed novel impacts of SMC-MR deletion on the cardiac cellulome in obese mice. Specifically, WD feeding induced inflammatory gene signatures in non-myocyte populations including B/T cells, macrophages, and endothelium as well as increased coronary VCAM-1 protein expression, independent of cardiac fibrosis, that was prevented by SMC-MR deletion. Further, SMC-MR deletion induced a basal reduction in cardiac mast cells and prevented WD-induced cardiac pro-inflammatory chemokine expression and leukocyte recruitment. These data reveal a central role for SMC-MR signaling in obesity-associated coronary and cardiac dysfunction, thus supporting the emerging paradigm of a vascular origin of cardiac dysfunction in obesity.
Collapse
Affiliation(s)
- Malathi S I Dona
- Baker Heart and Diabetes Research Institute, 75 Commercial Rd Prahran, Melbourne, VIC, 3004, Australia
| | - Ian Hsu
- Baker Heart and Diabetes Research Institute, 75 Commercial Rd Prahran, Melbourne, VIC, 3004, Australia
| | - Alex I Meuth
- Biomedical Sciences, University of Missouri, E102 Vet Med Bldg, Columbia, MO, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Scott M Brown
- Biomedical Sciences, University of Missouri, E102 Vet Med Bldg, Columbia, MO, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Chastidy A Bailey
- Biomedical Sciences, University of Missouri, E102 Vet Med Bldg, Columbia, MO, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Christian G Aragonez
- Biomedical Sciences, University of Missouri, E102 Vet Med Bldg, Columbia, MO, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Jacob J Russell
- Biomedical Sciences, University of Missouri, E102 Vet Med Bldg, Columbia, MO, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Crisdion Krstevski
- Baker Heart and Diabetes Research Institute, 75 Commercial Rd Prahran, Melbourne, VIC, 3004, Australia
| | - Annayya R Aroor
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
- Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Bysani Chandrasekar
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
- Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Vincent G DeMarco
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA
- Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Laurel A Grisanti
- Biomedical Sciences, University of Missouri, E102 Vet Med Bldg, Columbia, MO, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Alexander R Pinto
- Baker Heart and Diabetes Research Institute, 75 Commercial Rd Prahran, Melbourne, VIC, 3004, Australia.
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia.
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, E102 Vet Med Bldg, Columbia, MO, USA.
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.
- Research Service, Harry S Truman Memorial Veterans Hospital, Columbia, MO, USA.
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Endothelial dysfunction is a major risk factor for many cardiovascular diseases, notably hypertension. Obesity increases the risk of endothelial dysfunction in association with increasing production of the adipokine leptin. Preclinical studies have begun to unravel the mechanisms whereby leptin leads to the development of endothelial dysfunction, which are sex-specific. This review will summarize recent findings of mechanisms of leptin-induced endothelial impairment in both male and females and in pregnancy. RECENT FINDINGS Leptin receptors are found in high concentrations in the central nervous system (CNS), via which leptin promotes appetite suppression and upregulates sympathetic nervous system activation. However, leptin receptors are expressed in many other tissues, including the vascular endothelial cells and smooth muscle cells. Recent studies in mice with vascular endothelial or smooth muscle-specific knockdown demonstrate that endothelial leptin receptor activation plays a protective role against endothelial dysfunction in male animals, but not necessarily in females. Clinical studies indicate that women may be more sensitive to obesity-associated vascular endothelial dysfunction. Emerging preclinical data indicates that leptin and progesterone increase aldosterone production and endothelial mineralocorticoid receptor activation, respectively. Furthermore, decades of clinical studies indicate that leptin levels increase in the hypertensive pregnancy disorder preeclampsia, which is characterized by systemic endothelial dysfunction. Leptin infusion in mice induces the clinical characteristics of preeclampsia, including endothelial dysfunction. SUMMARY Novel preclinical data indicate that the mechanisms whereby leptin promotes endothelial dysfunction are sex-specific. Leptin-induced endothelial dysfunction may also play a role in hypertensive pregnancy as well.
Collapse
Affiliation(s)
- Elisabeth Mellott
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA
| | - Jessica L Faulkner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA
- Department of OBGYN, Medical College of Georgia at Augusta University, Augusta, GA
| |
Collapse
|
14
|
Mamazhakypov A, Lother A. Therapeutic targeting of mineralocorticoid receptors in pulmonary hypertension: Insights from basic research. Front Cardiovasc Med 2023; 10:1118516. [PMID: 36793473 PMCID: PMC9922727 DOI: 10.3389/fcvm.2023.1118516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Pulmonary hypertension (PH) is characterized by pulmonary vascular remodeling and associated with adverse outcomes. In patients with PH, plasma aldosterone levels are elevated, suggesting that aldosterone and its receptor, the mineralocorticoid receptor (MR), play an important role in the pathophysiology of PH. The MR plays a crucial role in adverse cardiac remodeling in left heart failure. A series of experimental studies from the past few years indicate that MR activation promotes adverse cellular processes that lead to pulmonary vascular remodeling, including endothelial cell apoptosis, smooth muscle cell (SMC) proliferation, pulmonary vascular fibrosis, and inflammation. Accordingly, in vivo studies have demonstrated that pharmacological inhibition or cell-specific deletion of the MR can prevent disease progression and partially reverse established PH phenotypes. In this review, we summarize recent advances in MR signaling in pulmonary vascular remodeling based on preclinical research and discuss the potential, but also the challenges, in bringing MR antagonists (MRAs) into clinical application.
Collapse
Affiliation(s)
- Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Achim Lother
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany,Faculty of Medicine, Interdisciplinary Medical Intensive Care, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany,*Correspondence: Achim Lother,
| |
Collapse
|
15
|
Cardiovascular Disease in Obstructive Sleep Apnea: Putative Contributions of Mineralocorticoid Receptors. Int J Mol Sci 2023; 24:ijms24032245. [PMID: 36768567 PMCID: PMC9916750 DOI: 10.3390/ijms24032245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic and highly prevalent condition that is associated with oxidative stress, inflammation, and fibrosis, leading to endothelial dysfunction, arterial stiffness, and vascular insulin resistance, resulting in increased cardiovascular disease and overall mortality rates. To date, OSA remains vastly underdiagnosed and undertreated, with conventional treatments yielding relatively discouraging results for improving cardiovascular outcomes in OSA patients. As such, a better mechanistic understanding of OSA-associated cardiovascular disease (CVD) and the development of novel adjuvant therapeutic targets are critically needed. It is well-established that inappropriate mineralocorticoid receptor (MR) activation in cardiovascular tissues plays a causal role in a multitude of CVD states. Clinical studies and experimental models of OSA lead to increased secretion of the MR ligand aldosterone and excessive MR activation. Furthermore, MR activation has been associated with worsened OSA prognosis. Despite these documented relationships, there have been no studies exploring the causal involvement of MR signaling in OSA-associated CVD. Further, scarce clinical studies have exclusively assessed the beneficial role of MR antagonists for the treatment of systemic hypertension commonly associated with OSA. Here, we provide a comprehensive overview of overlapping mechanistic pathways recruited in the context of MR activation- and OSA-induced CVD and propose MR-targeted therapy as a potential avenue to abrogate the deleterious cardiovascular consequences of OSA.
Collapse
|
16
|
Wolter NL, Jaffe IZ. Emerging vascular cell-specific roles for mineralocorticoid receptor: implications for understanding sex differences in cardiovascular disease. Am J Physiol Cell Physiol 2023; 324:C193-C204. [PMID: 36440858 PMCID: PMC9902217 DOI: 10.1152/ajpcell.00372.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
As growing evidence implicates extrarenal mineralocorticoid receptor (MR) in cardiovascular disease (CVD), recent studies have defined both cell- and sex-specific roles. MR is expressed in vascular smooth muscle (SMC) and endothelial cells (ECs). This review integrates published data from the past 5 years to identify novel roles for vascular MR in CVD, with a focus on understanding sex differences. Four areas are reviewed in which there is recently expanded understanding of the cell type- or sex-specific role of MR in 1) obesity-induced microvascular endothelial dysfunction, 2) vascular inflammation in atherosclerosis, 3) pulmonary hypertension, and 4) chronic kidney disease (CKD)-related CVD. The review focuses on preclinical data on each topic describing new mechanistic paradigms, cell type-specific mechanisms, sexual dimorphism if addressed, and clinical implications are then considered. New data support that MR drives vascular dysfunction induced by cardiovascular risk factors via sexually dimorphic mechanisms. In females, EC-MR contributes to obesity-induced endothelial dysfunction by regulating epithelial sodium channel expression and by inhibiting estrogen-induced nitric oxide production. In males with hyperlipidemia, EC-MR promotes large vessel inflammation by genomic regulation of leukocyte adhesion molecules, which is inhibited by the estrogen receptor. In pulmonary hypertension models, MRs in EC and SMC contribute to distinct components of disease pathologies including pulmonary vessel remodeling and RV dysfunction. Despite a female predominance in pulmonary hypertension, sex-specific roles for MR have not been explored. Vascular MR has also been directly implicated in CKD-related vascular dysfunction, independent of blood pressure. Despite these advances, sex differences in MR function remain understudied.
Collapse
Affiliation(s)
- Nicole L Wolter
- Molecular Cardiology Research Institute, https://ror.org/002hsbm82Tufts Medical Center, Boston, Massachusetts
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, https://ror.org/002hsbm82Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
17
|
Ibarrola J, Lu Q, Zennaro MC, Jaffe IZ. Mechanism by Which Inflammation and Oxidative Stress Induce Mineralocorticoid Receptor Gene Expression in Aging Vascular Smooth Muscle Cells. Hypertension 2023; 80:111-124. [PMID: 36337050 PMCID: PMC9742321 DOI: 10.1161/hypertensionaha.122.19213] [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: 02/16/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Vascular MR (mineralocorticoid receptor) expression increases with age driving aging-associated vascular stiffness and hypertension. MR has two isoforms (1α and 1β) with distinct 5'-untranslated and promoter sequences (P1 and P2), but the gene regulatory mechanisms remain unknown. We investigated mechanisms driving MR gene transcriptional regulation in aging human smooth muscle cells (SMC). METHODS MR was quantified in aortic tissue and primary human aortic SMC (HASMC) comparing adult and aged donors and adult HASMC treated with H2O2, to induce aging. Predicted transcription factor (TF) binding sites in the MR gene were validated using chromatin immunoprecipitations and reporter assays. The impact of TF inhibitors on MR isoforms and fibrosis target gene expression was examined. RESULTS Expression of both MR mRNA isoforms increased with donor age or H2O2 treatment in HASMCs. HIF1α (hypoxia-inducible factor) and the inflammatory TF NFκB (nuclear factor kappa B) both increased with age in HASMCs and are predicted to bind MR promoters. H2O2 induced HIF1α and NFκB expression and DNA binding of HIF1α to the MR P1 promoter and of NFκB to both MR promoters in HASMCs. HIF1α inhibition decreased MR-1α isoform expression while NFκB inhibition decreased both MR isoforms. HIF1α, NFκB, and MR inhibition decreased the expression of a SMC-MR target gene implicated in vascular fibrosis. In human aortic tissues, expression of HIF1α and NFκB each positively correlated with donor age and MR expression (P<0.0001). CONCLUSIONS These data implicate the inflammatory TF, NFκB, and oxidative stress-induced TF, HIF1α, in regulating SMC MR transcription in aging HASMCs, which drives aging-related vascular stiffness and cardiovascular disease.
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | | | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| |
Collapse
|
18
|
Abstract
Besides the physiological regulation of water, sodium, and potassium homeostasis, aldosterone modulates several physiological and pathological processes in the cardiovascular system. At the vascular level, aldosterone excess stimulates endothelial dysfunction and infiltration of inflammatory cells, enhances the development of the atherosclerotic plaque, and favors plaque instability, arterial stiffness, and calcification. At the cardiac level, aldosterone increases cardiac inflammation, fibrosis, and myocardial hypertrophy. As a clinical consequence, high aldosterone levels are associated with enhanced risk of cardiovascular events and mortality, especially when aldosterone secretion is inappropriate for renin levels and sodium intake, as in primary aldosteronism. Several clinical trials showed that mineralocorticoid receptor antagonists reduce cardiovascular mortality in patients with heart failure and reduced ejection fraction, but inconclusive results were reported for other cardiovascular conditions, such as heart failure with preserved ejection fraction, myocardial infarction, and atrial fibrillation. In patients with primary aldosteronism, adrenalectomy or treatment with mineralocorticoid receptor antagonists significantly mitigate adverse aldosterone effects, reducing the risk of cardiovascular events, mortality, and incident atrial fibrillation. In this review, we will summarize the major preclinical and clinical studies investigating the cardiovascular damage mediated by aldosterone and the protective effect of mineralocorticoid receptor antagonists for the reduction of cardiovascular risk in patients with cardiovascular diseases and primary aldosteronism.
Collapse
Affiliation(s)
- Fabrizio Buffolo
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Martina Tetti
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| |
Collapse
|
19
|
Li H, Konja D, Wang L, Wang Y. Sex Differences in Adiposity and Cardiovascular Diseases. Int J Mol Sci 2022; 23:ijms23169338. [PMID: 36012601 PMCID: PMC9409326 DOI: 10.3390/ijms23169338] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
Body fat distribution is a well-established predictor of adverse medical outcomes, independent of overall adiposity. Studying body fat distribution sheds insights into the causes of obesity and provides valuable information about the development of various comorbidities. Compared to total adiposity, body fat distribution is more closely associated with risks of cardiovascular diseases. The present review specifically focuses on the sexual dimorphism in body fat distribution, the biological clues, as well as the genetic traits that are distinct from overall obesity. Understanding the sex determinations on body fat distribution and adiposity will aid in the improvement of the prevention and treatment of cardiovascular diseases (CVD).
Collapse
|
20
|
Igbekele AE, Jia G, Hill MA, Sowers JR, Jia G. Mineralocorticoid Receptor Activation in Vascular Insulin Resistance and Dysfunction. Int J Mol Sci 2022; 23:8954. [PMID: 36012219 PMCID: PMC9409140 DOI: 10.3390/ijms23168954] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022] Open
Abstract
Systemic insulin resistance is characterized by reduced insulin metabolic signaling and glucose intolerance. Mineralocorticoid receptors (MRs), the principal receptors for the hormone aldosterone, play an important role in regulating renal sodium handling and blood pressure. Recent studies suggest that MRs also exist in tissues outside the kidney, including vascular endothelial cells, smooth muscle cells, fibroblasts, perivascular adipose tissue, and immune cells. Risk factors, including excessive salt intake/salt sensitivity, hypertension, and obesity, can lead to the activation of vascular MRs to promote inflammation, oxidative stress, remodeling, and fibrosis, as well as cardiovascular stiffening and microcirculatory impairment. These pathophysiological changes are associated with a diminished ability of insulin to initiate appropriate intracellular signaling events, resulting in a reduced glucose uptake within the microcirculation and related vascular insulin resistance. Therefore, the pharmacological inhibition of MR activation provides a potential therapeutic option for improving vascular function, glucose uptake, and vascular insulin sensitivity. This review highlights recent experimental and clinical data that support the contribution of abnormal MR activation to the development of vascular insulin resistance and dysfunction.
Collapse
Affiliation(s)
- Aderonke E. Igbekele
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - George Jia
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Michael A. Hill
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65212, USA
| | - James R. Sowers
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65212, USA
| | - Guanghong Jia
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
21
|
Yu S, Cui K, Wu P, Wu B, Lu X, Huang R, Tang X, Lin J, Yang B, Zhao J, He Q, Liang X, Xu Y. Melatonin prevents experimental central serous chorioretinopathy in rats. J Pineal Res 2022; 73:e12802. [PMID: 35436360 DOI: 10.1111/jpi.12802] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/28/2022]
Abstract
Central serous chorioretinopathy (CSC) is a vision-threatening disease with no validated treatment and unclear pathogenesis. It is characterized by dilation and leakage of choroidal vasculature, resulting in the accumulation of subretinal fluid, and serous detachment of the neurosensory retina. Numerous studies have demonstrated that melatonin had multiple protective effects against endothelial dysfunction, vascular inflammation, and blood-retinal barrier (BRB) breakdown. However, the effect of melatonin on CSC, and its exact pathogenesis, is not well understood thus far. In this study, an experimental model was established by intravitreal injection of aldosterone in rats, which mimicked the features of CSC. Our results found that melatonin administration in advance significantly inhibited aldosterone-induced choroidal thickening and vasodilation by reducing the expression of calcium-activated potassium channel KCa2.3, and attenuated tortuosity of choroid vessels. Moreover, melatonin protected the BRB integrity and prevented the decrease in tight junction protein (ZO-1, occludin, and claudin-1) levels in the rat model induced by aldosterone. Additionally, the data also showed that intraperitoneal injection of melatonin in advance inhibited aldosterone-induced macrophage/microglia infiltration, and remarkably diminished the levels of inflammatory cytokines (interleukin-6 [IL-6], IL-1β, and cyclooxygenase-2), chemokines (chemokine C-C motif ligand 3, and C-X-C motif ligand 1), and matrix metalloproteinases (MMP-2 and MMP-9). Luzindole, as the nonselective MT1 and MT2 antagonist, and 4-phenyl-2-propionamidotetraline, as the selective MT2 antagonist, neutralized the melatonin-induced inhibition of choroidal thickening and choroidal vasodilation, indicating that melatonin might exert the effects via binding to its receptors. Furthermore, the IL-17A/nuclear factor-κB signaling pathway was activated by intravitreal administration of aldosterone, while it was suppressed in melatonin-treated in advance rat eyes. This study indicates that melatonin could serve as a promising safe therapeutic strategy for CSC patients.
Collapse
Affiliation(s)
- Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Peiqi Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Benjuan Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Rong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jianqiang Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jinfeng Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qingjing He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
22
|
Faulkner JL, Wright D, Antonova G, Jaffe IZ, Kennard S, Belin de Chantemèle EJ. Midgestation Leptin Infusion Induces Characteristics of Clinical Preeclampsia in Mice, Which Is Ablated by Endothelial Mineralocorticoid Receptor Deletion. Hypertension 2022; 79:1536-1547. [PMID: 35510543 DOI: 10.1161/hypertensionaha.121.18832] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Patients with preeclampsia demonstrate increases in placental leptin production in midgestation, and an associated increase in late gestation plasma leptin levels. The consequences of mid-late gestation increases in leptin production in pregnancy is unknown. Our previous work indicates that leptin infusion induces endothelial dysfunction in nonpregnant female mice via leptin-mediated aldosterone production and endothelial mineralocorticoid receptor (ECMR) activation, which is ablated by ECMR deletion. Therefore, we hypothesized that leptin infusion in mid-gestation of pregnancy induces endothelial dysfunction and hypertension, hallmarks of clinical preeclampsia, which are prevented by ECMR deletion. METHODS Leptin was infused via miniosmotic pump (0.9 mg/kg per day) into timed-pregnant ECMR-intact (WT) and littermate-mice with ECMR deletion (KO) on gestation day (GD)11-18. RESULTS Leptin infusion decreased fetal weight and placental efficiency in WT mice compared with WT+vehicle. Radiotelemetry recording demonstrated that blood pressure increased in leptin-infused WT mice during infusion. Leptin infusion reduced endothelial-dependent relaxation responses to acetylcholine (ACh) in both resistance (second-order mesenteric) and conduit (aorta) vessels in WT pregnant mice. Leptin infusion increased placental ET-1 (endothelin-1) production evidenced by increased PPET-1 (preproendothelin-1) and ECE-1 (endothelin-converting enzyme-1) expressions in WT mice. Adrenal aldosterone synthase (CYP11B2) and angiotensin II type 1 receptor b (AT1Rb) expression increased with leptin infusion in pregnant WT mice. KO pregnant mice demonstrated protection from leptin-induced reductions in pup weight, placental efficiency, increased BP, and endothelial dysfunction. CONCLUSIONS Collectively, these data indicate that leptin infusion in midgestation induces endothelial dysfunction, hypertension, and fetal growth restriction in pregnant mice, which is ablated by ECMR deletion.
Collapse
Affiliation(s)
- Jessica L Faulkner
- Department of Physiology (J.L.F.), Medical College of Georgia at Augusta University
| | - Derrian Wright
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Galina Antonova
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (I.Z.J.)
| | - Simone Kennard
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University
| | - Eric J Belin de Chantemèle
- Vascular Biology Center (D.W., G.A., S.K., E.J.B.d.C.), Medical College of Georgia at Augusta University.,Department of Cardiology (E.J.B.d.C.), Medical College of Georgia at Augusta University
| |
Collapse
|
23
|
Sabe SA, Feng J, Sellke FW, Abid MR. Mechanisms and clinical implications of endothelium-dependent vasomotor dysfunction in coronary microvasculature. Am J Physiol Heart Circ Physiol 2022; 322:H819-H841. [PMID: 35333122 PMCID: PMC9018047 DOI: 10.1152/ajpheart.00603.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022]
Abstract
Coronary microvascular disease (CMD), which affects the arterioles and capillary endothelium that regulate myocardial perfusion, is an increasingly recognized source of morbidity and mortality, particularly in the setting of metabolic syndrome. The coronary endothelium plays a pivotal role in maintaining homeostasis, though factors such as diabetes, hypertension, hyperlipidemia, and obesity can contribute to endothelial injury and consequently arteriolar vasomotor dysfunction. These disturbances in the coronary microvasculature clinically manifest as diminished coronary flow reserve, which is a known independent risk factor for cardiac death, even in the absence of macrovascular atherosclerotic disease. Therefore, a growing body of literature has examined the molecular mechanisms by which coronary microvascular injury occurs at the level of the endothelium and the consequences on arteriolar vasomotor responses. This review will begin with an overview of normal coronary microvascular physiology, modalities of measuring coronary microvascular function, and clinical implications of CMD. These introductory topics will be followed by a discussion of recent advances in the understanding of the mechanisms by which inflammation, oxidative stress, insulin resistance, hyperlipidemia, hypertension, shear stress, endothelial cell senescence, and tissue ischemia dysregulate coronary endothelial homeostasis and arteriolar vasomotor function.
Collapse
Affiliation(s)
- Sharif A Sabe
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jun Feng
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
24
|
Barrera-Chimal J, Bonnard B, Jaisser F. Roles of Mineralocorticoid Receptors in Cardiovascular and Cardiorenal Diseases. Annu Rev Physiol 2022; 84:585-610. [PMID: 35143332 DOI: 10.1146/annurev-physiol-060821-013950] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mineralocorticoid receptor (MR) activation in the heart and vessels leads to pathological effects, such as excessive extracellular matrix accumulation, oxidative stress, and sustained inflammation. In these organs, the MR is expressed in cardiomyocytes, fibroblasts, endothelial cells, smooth muscle cells, and inflammatory cells. We review the accumulating experimental and clinical evidence that pharmacological MR antagonism has a positive impact on a battery of cardiac and vascular pathological states, including heart failure, myocardial infarction, arrhythmic diseases, atherosclerosis, vascular stiffness, and cardiac and vascular injury linked to metabolic comorbidities and chronic kidney disease. Moreover, we present perspectives on optimization of the use of MR antagonists in patients more likely to respond to such therapy and review the evidence suggesting that novel nonsteroidal MR antagonists offer an improved safety profile while retaining their cardiovascular protective effects. Finally, we highlight future therapeutic applications of MR antagonists in cardiovascular injury.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Benjamin Bonnard
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France;
| | - Frederic Jaisser
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France; .,INSERM Centre d'Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN INI-CRCT), Université de Lorraine, Nancy, France
| |
Collapse
|
25
|
Lyngsø KS, Jensen BL, Hansen PBL, Dimke H. Endothelial mineralocorticoid receptor ablation confers protection towards endothelial dysfunction in experimental diabetes in mice. Acta Physiol (Oxf) 2022; 234:e13731. [PMID: 34519423 DOI: 10.1111/apha.13731] [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: 02/05/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 11/29/2022]
Abstract
AIM With diabetes comes a significant risk of macrovascular and microvascular complications. Circulating aldosterone levels increase in patients with diabetes. Aldosterone can directly affect vascular function via activation of the mineralocorticoid receptor (MR). We hypothesized that aldosterone via endothelial MR impairs endothelial function in a murine model of experimental diabetes. METHOD Endothelial cell-specific mineralocorticoid receptor knockout MRflox/flox ; Tie2-Cre mice (ECMR-KO) and wild-type FVB littermates were subjected to an experimental type-1 diabetic model by low dose streptozotocin injections (55mg/kg/day) for five consecutive days. After 10 weeks of diabetes, second-order mesenteric resistance arteries were perfused ex vivo to evaluate vessel contractility and endothelial function. The effect of ex vivo incubation with aldosterone with and without the antagonist, spironolactone was determined. RESULTS Diabetic ECMR-KO and wild-type mice had similar, elevated, plasma aldosterone concentration while only diabetic wild-type mice displayed elevated urine albumin excretion and cardiac and kidney hypertrophy at 10 weeks. There were no differences in contraction (Emax and EC50 ) to thromboxane receptor agonist (U46619) and elevated K+ between groups. Wild-type diabetic mice showed impaired acetylcholine (ACh)-dependent relaxation, while diabetic ECMR-KO mice had intact ACh-mediated relaxation. Aldosterone incubation ex vivo impaired ACh mediated relaxation and rendered responses similar to diabetic WT arteries. Direct, ex vivo aldosterone effects were absent in ECMR-KO animals. Ex vivo inhibitory effects of aldosterone on endothelial relaxation in arteries from WT were abolished by spironolactone. CONCLUSION These findings show that endothelial cell mineralocorticoid receptor activation accounts for diabetes-induced systemic endothelial dysfunction in experimental diabetes and may explain the cardiovascular protection by MR antagonists in diabetes.
Collapse
Affiliation(s)
- Kristina S. Lyngsø
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense C Denmark
| | - Boye L. Jensen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense C Denmark
| | - Pernille B. L. Hansen
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense C Denmark
- Bioscience Renal, Research and Early Development Cardiovascular, Renal and Metabolism BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research Institute of Molecular Medicine University of Southern Denmark Odense C Denmark
- Department of Nephrology Odense University Hospital Odense Denmark
| |
Collapse
|
26
|
Bauersachs J, López-Andrés N. Mineralocorticoid receptor in cardiovascular diseases-Clinical trials and mechanistic insights. Br J Pharmacol 2021; 179:3119-3134. [PMID: 34643952 DOI: 10.1111/bph.15708] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/07/2021] [Accepted: 09/27/2021] [Indexed: 12/19/2022] Open
Abstract
Aldosterone binds to the mineralocorticoid receptor (NR3C2), a transcription factor of the nuclear receptor family, present in the kidney and in various other non-epithelial cells including the heart and the vasculature. Indeed, extra-renal pathophysiological effects of this hormone have been characterized, extending its actions to the cardiovascular system. A growing body of clinical and pre-clinical evidence suggests that mineralocorticoid receptor overactivation plays an important pathophysiological role in cardiovascular remodelling by promoting cardiac hypertrophy, fibrosis, arterial stiffness and in inflammation and oxidative stress. The following review article outlines the role of mineralocorticoid receptor in cardiovascular disease with a focus on myocardial remodelling and heart failure (HF) including clinical trials as well as cellular and animal studies.
Collapse
Affiliation(s)
- Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Natalia López-Andrés
- Cardiovascular Translational Research. Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| |
Collapse
|
27
|
Victorio JA, Guizoni DM, Freitas IN, Araujo TR, Davel AP. Effects of High-Fat and High-Fat/High-Sucrose Diet-Induced Obesity on PVAT Modulation of Vascular Function in Male and Female Mice. Front Pharmacol 2021; 12:720224. [PMID: 34566644 PMCID: PMC8460896 DOI: 10.3389/fphar.2021.720224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Increased adiposity in perivascular adipose tissue (PVAT) has been related to vascular dysfunction. High-fat (HF) diet-induced obesity models are often used to analyze the translational impact of obesity, but differences in sex and Western diet type complicate comparisons between studies. The role of PVAT was investigated in small mesenteric arteries (SMAs) of male and female mice fed a HF or a HF plus high-sucrose (HF + HS) diet for 3 or 5 months and compared them to age/sex-matched mice fed a chow diet. Vascular responses of SMAs without (PVAT-) or with PVAT (PVAT+) were evaluated. HF and HF + HS diets increased body weight, adiposity, and fasting glucose and insulin levels without affecting blood pressure and circulating adiponectin levels in both sexes. HF or HF + HS diet impaired PVAT anticontractile effects in SMAs from females but not males. PVAT-mediated endothelial dysfunction in SMAs from female mice after 3 months of a HF + HS diet, whereas in males, this effect was observed only after 5 months of HF + HS diet. However, PVAT did not impact acetylcholine-induced relaxation in SMAs from both sexes fed HF diet. The findings suggest that the addition of sucrose to a HF diet accelerates PVAT dysfunction in both sexes. PVAT dysfunction in response to both diets was observed early in females compared to age-matched males suggesting a susceptibility of the female sex to PVAT-mediated vascular complications in the setting of obesity. The data illustrate the importance of the duration and composition of obesogenic diets for investigating sex-specific treatments and pharmacological targets for obesity-induced vascular complications.
Collapse
Affiliation(s)
- Jamaira A Victorio
- Department of Structural and Functional Biology, Laboratory of Vascular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Daniele M Guizoni
- Department of Structural and Functional Biology, Laboratory of Vascular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Israelle N Freitas
- Department of Structural and Functional Biology, Laboratory of Vascular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Thiago R Araujo
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center-OCRC, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Ana P Davel
- Department of Structural and Functional Biology, Laboratory of Vascular Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|
28
|
Zhang S, Yang F, Shi R, Liu C, Zhao L, Gu X, Liu Y, Fu F, Feng N, Liu Y, Jia M, Fan R, Yang L, Li J, Li J, Pei J. Activation of κ-opioid receptor inhibits inflammatory response induced by sodium palmitate in human umbilical vein endothelial cells. Cytokine 2021; 146:155659. [PMID: 34332276 DOI: 10.1016/j.cyto.2021.155659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVES The current study aims to investigate the effect of κ-opioid receptor (κ-OR) activation on sodium palmitate (SP)-induced human umbilical vein endothelial cells (HUVECs) inflammatory response and elucidate the underlying mechanisms. METHODS A hyperlipidemic cell model was established and treated with κ-OR agonist (U50,488H), and antagonist (norbinaltorphimine, nor-BNI), or inhibitors targeting PI3K, Akt or eNOS (LY294002, MK2206-2HCl or L-NAME, respectively). Furthermore, the expression levels of NLRP3, caspase-1, p-Akt, Akt, p-eNOS, and total eNOS were evaluated. Additionally, the production of reactive oxygen species, and levels of inflammatory factors, such as TNF-α, IL-1β, IL-6, IL-1 and adhesion molecules, such as ICAM-1, VCAM-1, P-selectin, and E-selectin were determined. The adherence rates of the neutrophils and monocytes were assessed as well. RESULTS The SP-induced hyperlipidemic cell model demonstrated increased expression of NLRP3 and caspase-1 proteins (P < 0.05) and elevated ROS levels (P < 0.01), and decreased phosphorylated-Akt and phosphorylated-eNOS expression (P < 0.05). In addition, SP significantly increased TNF-α, IL-1β, IL-6, ICAM-1, VCAM-1, P-selectin, and E-selectin levels (P < 0.01), decreased IL-10 levels (P < 0.01), and increased the adhesion rates of monocytes and neutrophils (P < 0.01). The SP-induced inflammatory response in HUVECs was ameliorated by κ-OR agonist, U50,488H. However, the protective effect of U50,488H was abolished by κ-OR antagonist, nor-BNI, and inhibitors of PI3K, Akt and eNOS. CONCLUSION Our findings suggest that κ-OR activation inhibits SP-induced inflammation by activating the PI3K/Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Shumiao Zhang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Fan Yang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Rui Shi
- School of Life Sciences, Northwest University, No.1 North Taibai Road, Xi'an 710069, Shaanxi Province, People's Republic of China
| | - Chaoyang Liu
- School of Life Sciences, Northwest University, No.1 North Taibai Road, Xi'an 710069, Shaanxi Province, People's Republic of China
| | - Lei Zhao
- Department of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi'an 710033, Shaanxi Province, China
| | - Xiaoming Gu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Yinji Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Feng Fu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Na Feng
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Yali Liu
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Min Jia
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Rong Fan
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Lu Yang
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Jun Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China
| | - Juan Li
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China.
| | - Jianming Pei
- Department of Physiology and Pathophysiology, National Key Discipline of Cell Biology, Air Force Medical University, No. 169 West Changle Road, Xi'an 710032, Shaanxi Province, People's Republic of China; School of Life Sciences, Northwest University, No.1 North Taibai Road, Xi'an 710069, Shaanxi Province, People's Republic of China.
| |
Collapse
|
29
|
Menon DP, Qi G, Kim SK, Moss ME, Penumatsa KC, Warburton RR, Toksoz D, Wilson J, Hill NS, Jaffe IZ, Preston IR. Vascular cell-specific roles of mineralocorticoid receptors in pulmonary hypertension. Pulm Circ 2021; 11:20458940211025240. [PMID: 34211700 PMCID: PMC8216367 DOI: 10.1177/20458940211025240] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 05/13/2021] [Indexed: 12/18/2022] Open
Abstract
Abnormalities that characterize pulmonary arterial hypertension include impairment in the structure and function of pulmonary vascular endothelial and smooth muscle cells. Aldosterone levels are elevated in human pulmonary arterial hypertension and in experimental pulmonary hypertension, while inhibition of the aldosterone-binding mineralocorticoid receptor attenuates pulmonary hypertension in multiple animal models. We explored the role of mineralocorticoid receptor in endothelial and smooth muscle cells in using cell-specific mineralocorticoid receptor knockout mice exposed to sugen/hypoxia-induced pulmonary hypertension. Treatment with the mineralocorticoid receptor inhibitor spironolactone significantly reduced right ventricular systolic pressure. However, this is not reproduced by selective mineralocorticoid receptor deletion in smooth muscle cells or endothelial cells. Similarly, spironolactone attenuated the increase in right ventricular cardiomyocyte area independent of vascular mineralocorticoid receptor with no effect on right ventricular weight or interstitial fibrosis. Right ventricular perivascular fibrosis was significantly decreased by spironolactone and this was reproduced by specific deletion of mineralocorticoid receptor from endothelial cells. Endothelial cell-mineralocorticoid receptor deletion attenuated the sugen/hypoxia-induced increase in the leukocyte-adhesion molecule, E-selectin, and collagen IIIA1 in the right ventricle. Spironolactone also significantly reduced pulmonary arteriolar muscularization, independent of endothelial cell-mineralocorticoid receptor or smooth muscle cell-mineralocorticoid receptor. Finally, the degree of pulmonary perivascular inflammation was attenuated by mineralocorticoid receptor antagonism and was fully reproduced by smooth muscle cell-specific mineralocorticoid receptor deletion. These studies demonstrate that in the sugen/hypoxia pulmonary hypertension model, systemic-mineralocorticoid receptor blockade significantly attenuates the disease and that mineralocorticoid receptor has cell-specific effects, with endothelial cell-mineralocorticoid receptor contributing to right ventricular perivascular fibrosis and smooth muscle cell-mineralocorticoid receptor participating in pulmonary vascular inflammation. As mineralocorticoid receptor antagonists are being investigated to treat pulmonary arterial hypertension, these findings support novel mechanisms and potential mineralocorticoid receptor targets that mediate therapeutic benefits in patients.
Collapse
Affiliation(s)
- Divya P. Menon
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| | - Guanming Qi
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| | - Seung K. Kim
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
- Department of Sports Science, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - M. Elizabeth Moss
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Krishna C. Penumatsa
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| | - Rod R. Warburton
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| | - Deniz Toksoz
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| | - Jamie Wilson
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| | - Nicholas S. Hill
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Ioana R. Preston
- Pulmonary, Critical Care and Sleep Division, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
30
|
Effect of Omega-3 Fatty Acid Alone and in Combination with Proprietary Chromium Complex on Endothelial Function in Subjects with Metabolic Syndrome: A Randomized, Double-Blind, Parallel-Group Clinical Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2972610. [PMID: 34257675 PMCID: PMC8253643 DOI: 10.1155/2021/2972610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 02/01/2021] [Accepted: 06/12/2021] [Indexed: 11/18/2022]
Abstract
Metabolic syndrome (MetS) represents a cluster of metabolic abnormalities that include hypertension, central obesity, insulin resistance, and dyslipidemia and is strongly associated with an increased risk of diabetes, cardiovascular diseases (CVD), and all-cause mortality. Early diagnosis is important to employ lifestyle and risk factor modification. Existing therapies are limited. Studies report positive effect of omega-3 fatty acids (ω-3FA) on symptoms of metabolic syndrome. The present study was undertaken to evaluate the effect of ω-3FA alone and in combination with proprietary chromium complex (PCC) on endothelial function in subjects with metabolic syndrome. In this randomized, double-blind, parallel-group study, subjects were enrolled into the study after ethics committee (EC) approval and informed consent. Eligible subjects were randomized to receive ω-3FA concentrate 2000 mg (Group A-18 subjects), ω-3FA concentrate 2000 mg + PCC200 mcg (Group B-19 subjects), and ω-3FA concentrate 2000 mg + PCC400 mcg (Group C-21 subjects) daily for 12 weeks. Endothelial dysfunction as measured by reflection index (RI), biomarkers of oxidative stress (NO, MDA, and glutathione), and inflammation (hsCRP, endothelin-1, ICAM-1, and VCAM-1) were evaluated at baseline, 4, and 12 weeks. Lipid-profile and platelet-aggregation tests were performed at baseline and 12 weeks. Adverse drug reactions were recorded. Compliance was assessed by pill count method. GraphPad Prism8 was used for statistical analysis. Significant changes were seen from 4 weeks onwards in all the parameters evaluated. Significant improvement in RI% (mean ± SD = -2.56 ± 0.77 to -3.27 ± 0.67-group A, -2.33 ± 0.76 to 4.72 ± 0.79-group B; -2.39 ± 1.13 to 6.46 ± 1.00-group C) was seen at 12 weeks. Significant improvement in biomarkers of oxidative stress and inflammation was seen with all the treatment groups. Similarly, significant improvement in lipid profile was seen in group B and group C, while group A showed change in HDL, VLDL, and TG. Group C demonstrated the best response in the parameters evaluated. Three patients in group C reported gastrointestinal adverse events, which resolved spontaneously; none stopped the therapy. So, the addition of PCC to ω-3FA may prove to have beneficial effect in reducing cardiovascular morbidity in MetS patients.
Collapse
|
31
|
Obesity-associated cardiovascular risk in women: hypertension and heart failure. Clin Sci (Lond) 2021; 135:1523-1544. [PMID: 34160010 DOI: 10.1042/cs20210384] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/14/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023]
Abstract
The pathogenesis of obesity-associated cardiovascular diseases begins long prior to the presentation of a cardiovascular event. In both men and women, cardiovascular events, and their associated hospitalizations and mortality, are often clinically predisposed by the presentation of a chronic cardiovascular risk factor. Obesity increases the risk of cardiovascular diseases in both sexes, however, the clinical prevalence of obesity, as well as its contribution to crucial cardiovascular risk factors is dependent on sex. The mechanisms via which obesity leads to cardiovascular risk is also discrepant in women between their premenopausal, pregnancy and postmenopausal phases of life. Emerging data indicate that at all reproductive statuses and ages, the presentation of a cardiovascular event in obese women is strongly associated with hypertension and its subsequent chronic risk factor, heart failure with preserved ejection fraction (HFpEF). In addition, emerging evidence indicates that obesity increases the risk of both hypertension and heart failure in pregnancy. This review will summarize clinical and experimental data on the female-specific prevalence and mechanisms of hypertension and heart failure in women across reproductive stages and highlight the particular risks in pregnancy as well as emerging data in a high-risk ethnicity in women of African ancestry (AA).
Collapse
|
32
|
Aravani D, Kassi E, Chatzigeorgiou A, Vakrou S. Cardiometabolic Syndrome: An Update on Available Mouse Models. Thromb Haemost 2021; 121:703-715. [PMID: 33280078 DOI: 10.1055/s-0040-1721388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiometabolic syndrome (CMS), a disease entity characterized by abdominal obesity, insulin resistance (IR), hypertension, and hyperlipidemia, is a global epidemic with approximately 25% prevalence in adults globally. CMS is associated with increased risk for cardiovascular disease (CVD) and development of diabetes. Due to its multifactorial etiology, the development of several animal models to simulate CMS has contributed significantly to the elucidation of the disease pathophysiology and the design of therapies. In this review we aimed to present the most common mouse models used in the research of CMS. We found that CMS can be induced either by genetic manipulation, leading to dyslipidemia, lipodystrophy, obesity and IR, or obesity and hypertension, or by administration of specific diets and drugs. In the last decade, the ob/ob and db/db mice were the most common obesity and IR models, whereas Ldlr-/- and Apoe-/- were widely used to induce hyperlipidemia. These mice have been used either as a single transgenic or combined with a different background with or without diet treatment. High-fat diet with modifications is the preferred protocol, generally leading to increased body weight, hyperlipidemia, and IR. A plethora of genetically engineered mouse models, diets, drugs, or synthetic compounds that are available have advanced the understanding of CMS. However, each researcher should carefully select the most appropriate model and validate its consistency. It is important to consider the differences between strains of the same animal species, different animals, and most importantly differences to human when translating results.
Collapse
Affiliation(s)
- Dimitra Aravani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Styliani Vakrou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Cardiology, "Laiko" General Hospital, Athens, Greece
| |
Collapse
|
33
|
Pieronne‐Deperrois M, Guéret A, Djerada Z, Crochemore C, Harouki N, Henry J, Dumesnil A, Larchevêque M, do Rego J, do Rego J, Nicol L, Richard V, Jaisser F, Kolkhof P, Mulder P, Monteil C, Ouvrard‐Pascaud A. Mineralocorticoid receptor blockade with finerenone improves heart function and exercise capacity in ovariectomized mice. ESC Heart Fail 2021; 8:1933-1943. [PMID: 33742556 PMCID: PMC8120350 DOI: 10.1002/ehf2.13219] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/27/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS In post-menopausal women, incidence of heart failure with preserved ejection fraction is higher than in men. Hormonal replacement therapies did not demonstrate benefits. We tested whether the non-steroidal mineralocorticoid receptor antagonist finerenone limits the progression of heart failure in ovariectomized (OVX) mice with metabolic disorders. METHODS AND RESULTS Ovariectomy was performed in 4-month-old mice, treated or not at 7 months old for 1 month with finerenone (Fine) 1 mg/kg/day. Left ventricular (LV) cardiac and coronary endothelial functions were assessed by echocardiography, catheterization, and myography. Blood pressure was measured by plethysmography. Insulin and glucose tolerance tests were performed. Exercise capacity and spontaneous activity were measured on treadmill and in combined indirect calorimetric cages equipped with voluntary running wheel. OVX mice presented LV diastolic dysfunction without modification of ejection fraction compared with controls (CTL), whereas finerenone improved LV filling pressure (LV end-diastolic pressure, mmHg: CTL 3.48 ± 0.41, OVX 6.17 ± 0.30**, OVX + Fine 3.65 ± 0.55† , **P < 0.01 vs. CTL, † P < 0.05 vs. OVX) and compliance (LV end-diastolic pressure-volume relation, mmHg/RVU: CTL 1.65 ± 0.42, OVX 4.77 ± 0.37***, OVX + Fine 2.87 ± 0.26†† , ***P < 0.001 vs. CTL, †† P < 0.01 vs. OVX). Acetylcholine-induced endothelial-dependent relaxation of coronary arteries was impaired in ovariectomized mice and improved by finerenone (relaxation, %: CTL 86 ± 8, OVX 38 ± 3**, OVX + Fine 83 ± 7†† , **P < 0.01 vs. CTL, †† P < 0.01 vs. OVX). Finerenone improved decreased ATP production by subsarcolemmal mitochondria after ovariectomy. Weight gain, increased blood pressure, and decreased insulin and glucose tolerance in OVX mice were improved by finerenone. The exercise capacity at race was diminished in untreated OVX mice only. Spontaneous activity measurements in ovariectomized mice showed decreased horizontal movements, reduced time spent in a running wheel, and reduced VO2 and VCO2 , all parameters improved by finerenone. CONCLUSIONS Finerenone improved cardiovascular dysfunction and exercise capacity after ovariectomy-induced LV diastolic dysfunction with preserved ejection fraction.
Collapse
Affiliation(s)
| | - Alexandre Guéret
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Zoubir Djerada
- Pharmacology DepartmentReims University HospitalReimsFrance
| | - Clément Crochemore
- EA4651 Toxemac‐ABTE, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Najah Harouki
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Jean‐Paul Henry
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Anaïs Dumesnil
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Marine Larchevêque
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Jean‐Claude do Rego
- SCAC Behavioral Analysis Platform, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Jean‐Luc do Rego
- SCAC Behavioral Analysis Platform, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Lionel Nicol
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Vincent Richard
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Frédéric Jaisser
- Inserm U1138, Cordeliers Institute, Paris‐VI UniversityParisFrance
| | | | - Paul Mulder
- Inserm U1096 ENVI, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | - Christelle Monteil
- EA4651 Toxemac‐ABTE, Rouen Medical School, UNIROUEN, Normandy UniversityRouenFrance
| | | |
Collapse
|
34
|
Mineralocorticoid receptor blockade normalizes coronary resistance in obese swine independent of functional alterations in K v channels. Basic Res Cardiol 2021; 116:35. [PMID: 34018061 DOI: 10.1007/s00395-021-00879-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
Impaired coronary microvascular function (e.g., reduced dilation and coronary flow reserve) predicts cardiac mortality in obesity, yet underlying mechanisms and potential therapeutic strategies remain poorly understood. Mineralocorticoid receptor (MR) antagonism improves coronary microvascular function in obese humans and animals. Whether MR blockade improves in vivo regulation of coronary flow, a process involving voltage-dependent K+ (Kv) channel activation, or reduces coronary structural remodeling in obesity is unclear. Thus, the goals of this investigation were to determine the effects of obesity on coronary responsiveness to reductions in arterial PO2 and potential involvement of Kv channels and whether the benefit of MR blockade involves improved coronary Kv function or altered passive structural properties of the coronary microcirculation. Hypoxemia increased coronary blood flow similarly in lean and obese swine; however, baseline coronary vascular resistance was significantly higher in obese swine. Inhibition of Kv channels reduced coronary blood flow and augmented coronary resistance under baseline conditions in lean but not obese swine and had no impact on hypoxemic coronary vasodilation. Chronic MR inhibition in obese swine normalized baseline coronary resistance, did not influence hypoxemic coronary vasodilation, and did not restore coronary Kv function (assessed in vivo, ex vivo, and via patch clamping). Lastly, MR blockade prevented obesity-associated coronary arteriolar stiffening independent of cardiac capillary density and changes in cardiac function. These data indicate that chronic MR inhibition prevents increased coronary resistance in obesity independent of Kv channel function and is associated with mitigation of obesity-mediated coronary arteriolar stiffening.
Collapse
|
35
|
Biwer LA, Carvajal BV, Lu Q, Man JJ, Jaffe IZ. Mineralocorticoid and Estrogen Receptors in Endothelial Cells Coordinately Regulate Microvascular Function in Obese Female Mice. Hypertension 2021; 77:2117-2126. [PMID: 33934622 DOI: 10.1161/hypertensionaha.120.16911] [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: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lauren A Biwer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (L.A.B., B.V.C., Q.L., J.J.M., I.Z.J.)
| | - Brigett V Carvajal
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (L.A.B., B.V.C., Q.L., J.J.M., I.Z.J.).,Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA (B.V.C., J.J.M., I.Z.J.)
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (L.A.B., B.V.C., Q.L., J.J.M., I.Z.J.)
| | - Joshua J Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (L.A.B., B.V.C., Q.L., J.J.M., I.Z.J.).,Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA (B.V.C., J.J.M., I.Z.J.)
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA (L.A.B., B.V.C., Q.L., J.J.M., I.Z.J.).,Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston MA (B.V.C., J.J.M., I.Z.J.)
| |
Collapse
|
36
|
Intrinsic exercise capacity induces divergent vascular plasticity via arachidonic acid-mediated inflammatory pathways in female rats. Vascul Pharmacol 2021; 140:106862. [PMID: 33872803 DOI: 10.1016/j.vph.2021.106862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/03/2021] [Accepted: 04/07/2021] [Indexed: 12/31/2022]
Abstract
Metabolic syndrome prevalence has increased among US adults, particularly among non-hispanic white and black women. Sedentary behavior often leads to chronic inflammation, a triggering factor of metabolic syndrome. Given that intrinsic exercise capacity is genetically inherited, we questioned if low-grade chronic inflammation would be present in a female rat model of low intrinsic exercise capacity-induced metabolic syndrome, while beneficial increase of resolution of inflammation would be present in a female rat model of high intrinsic exercise capacity. In the vascular system, two primary markers for inflammation and resolution of inflammation are cyclooxygenase (COX) and lipoxygenase (LOX), respectively. Our study focused on the novel hypothesis that untrained, inherited exercise capacity induces divergent vascular plasticity via changes in the delicate balance between COX and LOX inflammatory mediators. We used divergent rat strains with low (LCR) and high (HCR) aerobic running capacity. By using animals with contrasting intrinsic exercise capacities, it is possible to determine the exact triggers that lead to inherited vascular plasticity in female rats. We observed that female LCR displayed increased periovarian fat pad and body weight, which is congruent with their obesity-presenting phenotype. Furthermore, LCR presented with vascular hypocontractility and increased COX and LOX-derived pro-inflammatory factors. On the other hand, HCR presented with a "shutdown" of COX-induced vasoconstriction and enhanced resolution of inflammation to maintain vascular tone and homeostasis. In conclusion, LCR display low-grade chronic inflammation via increased COX activity. These results provide mechanistic clues as to why lower intrinsic aerobic capacity correlates with a predisposition to risk of vascular disease. Conversely, being born with higher intrinsic aerobic capacity is a significant factor for improved vascular physiology in female rats.
Collapse
|
37
|
Manrique-Acevedo C, Padilla J, Naz H, Woodford ML, Ghiarone T, Aroor AR, Hulse JL, Cabral-Amador FJ, Martinez-Diaz V, Hans CP, Whaley-Connell A, Martinez-Lemus LA, Lastra G. Mineralocorticoid Receptor in Myeloid Cells Mediates Angiotensin II-Induced Vascular Dysfunction in Female Mice. Front Physiol 2021; 12:588358. [PMID: 33854438 PMCID: PMC8039313 DOI: 10.3389/fphys.2021.588358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/17/2021] [Indexed: 11/13/2022] Open
Abstract
Enhanced mineralocorticoid receptor (MR) signaling is critical to the development of endothelial dysfunction and arterial stiffening. However, there is a lack of knowledge about the role of MR-induced adipose tissue inflammation in the genesis of vascular dysfunction in women. In this study, we hypothesize that MR activation in myeloid cells contributes to angiotensin II (Ang II)-induced aortic stiffening and endothelial dysfunction in females via increased pro-inflammatory (M1) macrophage polarization. Female mice lacking MR in myeloid cells (MyMRKO) were infused with Ang II (500 ng/kg/min) for 4 weeks. This was followed by determinations of aortic stiffness and vasomotor responses, as well as measurements of markers of inflammation and macrophage infiltration/polarization in different adipose tissue compartments. MyMRKO mice were protected against Ang II-induced aortic endothelial stiffening, as assessed via atomic force microscopy in aortic explants, and vasorelaxation dysfunction, as measured by aortic wire myography. In alignment, MyMRKO mice were protected against Ang II-induced macrophage infiltration and M1 polarization in visceral adipose tissue (VAT) and thoracic perivascular adipose tissue (tPVAT). Collectively, this study demonstrates a critical role of MR activation in myeloid cells in the pathogenesis of vascular dysfunction in females associated with pro-inflammatory macrophage polarization in VAT and tPVAT. Our data have potential clinical implications for the prevention and management of cardiovascular disease in women, who are disproportionally at higher risk for poor outcomes.
Collapse
Affiliation(s)
- Camila Manrique-Acevedo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Huma Naz
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States
| | - Makenzie L Woodford
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Annayya R Aroor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Jack L Hulse
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States
| | | | - Vanesa Martinez-Diaz
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Chetan P Hans
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Adam Whaley-Connell
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States.,Division of Nephrology and Hypertension, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO, United States.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Guido Lastra
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States
| |
Collapse
|
38
|
Guizoni DM, Freitas IN, Victorio JA, Possebom IR, Araujo TR, Carneiro EM, Davel AP. Taurine treatment reverses protein malnutrition-induced endothelial dysfunction of the pancreatic vasculature: The role of hydrogen sulfide. Metabolism 2021; 116:154701. [PMID: 33417894 DOI: 10.1016/j.metabol.2021.154701] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/11/2020] [Accepted: 12/31/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Protein malnutrition in childhood predisposes individuals to vascular and pancreatic endocrine dysfunction, thus increasing the risk of diabetes and hypertension. Because taurine may reduce cardiometabolic risk, we hypothesized that taurine treatment has a beneficial effect on the pancreatic vasculature during protein restriction. METHODS AND RESULTS Weaned mice were fed a normal or a low-protein diet and were treated with or without taurine for 3 months. The lieno-pancreatic artery (LPA) from low-protein diet-treated mice exhibited impaired endothelium-dependent relaxation to acetylcholine that was associated with decreased endothelium-derived hyperpolarization (EDH), hydrogen sulfide (H2S) production, and H2S-synthesizing CBS expression and impaired vasorelaxation to an H2S-donor, NaHS. These changes were prevented by taurine treatment. We compared the effects of taurine with the effects of the direct vasodilator hydralazine and found that both normalized blood pressure and the endothelial vasodilator function of the LPA in the mice fed a protein-restricted diet. However, only taurine restored the CBS expression in the LPA and insulin secretion in response to high glucose. The LPA supplies the pancreas and shares morphometry with the mesenteric resistance artery (MRA). However, in the MRA, low-protein diet-induced endothelial dysfunction is driven by impaired NOS-derived NO with no changes in H2S signaling. CONCLUSIONS The results suggest that taurine protects against protein malnutrition-induced endothelial dysfunction in the LPA by upregulating the CBS-H2S pathway. Considering the importance of the pancreatic vasculature for endocrine islet activity, taurine may be a potential therapy for the vascular and metabolic dysfunction associated with malnutrition and comorbidities.
Collapse
Affiliation(s)
- Daniele M Guizoni
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Israelle N Freitas
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Jamaira A Victorio
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Isabela R Possebom
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Thiago R Araujo
- Obesity and Comorbidities Research Center-OCRC, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Everardo M Carneiro
- Obesity and Comorbidities Research Center-OCRC, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Ana P Davel
- Laboratory of Vascular Biology, Institute of Biology, Department of Structural and Functional Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
39
|
Faulkner JL, Harwood D, Kennard S, Antonova G, Clere N, Belin de Chantemèle EJ. Dietary sodium restriction sex specifically impairs endothelial function via mineralocorticoid receptor-dependent reduction in NO bioavailability in Balb/C mice. Am J Physiol Heart Circ Physiol 2021; 320:H211-H220. [PMID: 33095056 PMCID: PMC7847080 DOI: 10.1152/ajpheart.00413.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
Recent findings from our group demonstrated that females exhibit higher endothelial mineralocorticoid receptor (MR) expression than males, which predisposes them to aldosterone-mediated endothelial dysfunction in the context of metabolic disorders. However, whether the endothelium of female mice presents a higher propensity to MR-mediated dysfunction than that of males in the absence of comorbidities remains unknown. We therefore sought to investigate whether increasing aldosterone production endogenously with sodium restriction impairs endothelial function in otherwise healthy female mice. We fed male and female Balb/C mice a normal (0.4% NaCl; NSD) or sodium-restricted diet (0.05% NaCl; SRD) for 4 wk. Females exhibited higher baseline endothelial function (relaxation to acetylcholine) and lower vascular contractility (constriction to phenylephrine, serotonin, and KCl). However, SRD impaired endothelial-dependent relaxation and increased vascular contractility in female mice, effectively ablating the baseline sex difference. Female sex also increased baseline adrenal CYP11B2 expression; however, SRD significantly enhanced CYP11B2 expression in male and female mice and ablated the sex difference. Nitric oxide synthase (NOS) inhibition with Nω-nitro-l-arginine methyl ester hydrochloride eliminated both sex as well as diet-induced differences in endothelial dysfunction. In accordance, females demonstrated higher vascular endothelial NOS expression at baseline, which SRD significantly decreased. In addition, SRD diminished vascular NOX4 expression in female mice only. MR blockade with spironolactone-protected female mice from decreases in endothelial-dependent relaxation but not increases in vascular contractility. Utilizing sodium restriction as a method to increase plasma aldosterone levels in healthy female mice, we demonstrated that female mice are more susceptible to vascular damage via MR activation in the vascular endothelium only.NEW & NOTEWORTHY Female sex confers improved endothelial relaxation and vascular constriction responses in female Balb/C mice compared with males under baseline conditions. Sodium restriction impairs endothelial function, which is nitric oxide dependent, and increases vascular contractility in association with reduced vascular endothelial nitric oxide synthase and NOX4 expression in female mice ablating the baseline sex difference. Mineralocorticoid receptor antagonism ablates sodium restriction-induced endothelial dysfunction, but not increased vascular contractility, in female mice.
Collapse
Affiliation(s)
- Jessica L Faulkner
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Daisy Harwood
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Simone Kennard
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Galina Antonova
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Nicolas Clere
- Micro and Nanomédecines Translationelles-MINT, UNIV Angers, INSERM U1066, CNRS UMR 6021, Université Bretagne Loire, Angers, France
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Cardiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
40
|
DuPont JJ, Kim SK, Kenney RM, Jaffe IZ. Sex differences in the time course and mechanisms of vascular and cardiac aging in mice: role of the smooth muscle cell mineralocorticoid receptor. Am J Physiol Heart Circ Physiol 2021; 320:H169-H180. [PMID: 33095647 PMCID: PMC7847078 DOI: 10.1152/ajpheart.00262.2020] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 10/16/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023]
Abstract
Aging is associated with heart and vascular dysfunction that contributes to cardiovascular disease (CVD) risk. Clinical data support a sexual dimorphism in the time course of aging-associated CVD. However, the mechanisms driving sex differences in cardiovascular aging and whether they can be modeled in mice have not been explored. Mineralocorticoid receptors (MRs) regulate blood pressure, and we previously demonstrated in male mice that MR expression increases in aging mouse vessels and smooth muscle cell-specific MR deletion (SMC-MR-KO) protects from cardiovascular aging. This study characterizes sex differences in murine cardiovascular aging and the associated sex-specific role of SMC-MR. Aortic stiffness, measured by pulse wave velocity, increased from 3 to 12 mo of age in males but not until 18 mo in females. The timing of the rise in aortic stiffening correlated with the timing of increased aortic MR expression, and aortic stiffness did not increase with age in SMC-MR-KO mice of both sexes. Vascular fibrosis increased at 12 mo in males and later at 18 mo in females; however, fibrosis was attenuated by SMC-MR-KO in males only. In resistance vessels, angiotensin type 1 receptor (AT1R)-mediated vasoconstriction also increased at 12 mo in males and 18 mo in females. ANG II-induced vasoconstriction was decreased in SMC-MR-KO specifically in males in association with decreased AT1R expression. Cardiac systolic function declined in males and females by 18 mo of age, which was prevented by SMC-MR-KO specifically in females. Cardiac perivascular fibrosis increased with age in both sexes accompanied by sex-specific changes in the expression levels of MR-regulated profibrotic genes.NEW & NOTEWORTHY These data demonstrate that the delayed and steeper decline in cardiovascular function observed in aging females can be modeled in aging mice. Moreover, the mechanisms driving vascular and cardiac aging phenotypes are distinct between males and females. Mineralocorticoid receptors in smooth muscle cells play a significant role in cardiovascular aging in both sexes; however, they do so by distinct mechanisms. Overall, these findings suggest that sex-specific therapies may be necessary to retard the aging process and improve cardiovascular disease outcomes in the aging population.
Collapse
Affiliation(s)
- Jennifer J DuPont
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Seung Kyum Kim
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
- Department of Sports Science, Seoul National University of Science and Technology, Seoul, South Korea
| | - Rachel M Kenney
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts
| |
Collapse
|
41
|
Faulkner JL, Lluch E, Kennard S, Antonova G, Jaffe IZ, Belin de Chantemèle EJ. Selective deletion of endothelial mineralocorticoid receptor protects from vascular dysfunction in sodium-restricted female mice. Biol Sex Differ 2020; 11:64. [PMID: 33228767 PMCID: PMC7685592 DOI: 10.1186/s13293-020-00340-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Background Recent evidence by our laboratory demonstrates that women and female mice endogenously express higher endothelial mineralocorticoid receptor (ECMR) than males. Mounting clinical evidence also indicates that aldosterone production is higher in pathological conditions in females compared to males. However, the role for increased activation of ECMR by aldosterone in the absence of a comorbid condition is yet to be explored. The current study hypothesized that increased ECMR activation induced by elevated aldosterone production predisposes healthy female mice to endothelial dysfunction. Method Vascular reactivity was assessed in aortic rings from wild-type (WT) and ECMR KO (KO) mice fed either a normal salt (NSD, 0.4% NaCl) or sodium-restricted diet (SRD, 0.05% NaCl) for 28 days. Results SRD elevated plasma aldosterone levels as well as adrenal CYP11B2 and angiotensin II type 1 receptor (AT1R) expressions in female, but not male, WT mice. In baseline conditions (NSD), endothelial function, assessed by vascular relaxation to acetylcholine, was higher while vascular contractility to phenylephrine, serotonin, and KCl lower in female than male WT mice. SRD impaired endothelial function and increased vascular contractility in female, but not male, WT mice effectively ablating the baseline sex differences. NOS inhibition with LNAME ablated endothelial relaxation to a higher extent in male than female mice on NSD and ablated differences in acetylcholine relaxation responses between NSD- and SRD-fed females, indicating a role for NO in SRD-mediated endothelial function. In association, SRD significantly reduced vascular NOX4 expression in female, but not male, mice. Lastly, selective deletion of ECMR protected female mice from SRD-mediated endothelial dysfunction and increased vascular contractility. Conclusion Collectively, these data indicate that female mice develop aldosterone-induced endothelial dysfunction via endothelial MR-mediated reductions in NO bioavailability. In addition, these data support a role for ECMR to promote vascular contractility in female mice in response to sodium restriction.
Collapse
Affiliation(s)
- Jessica L Faulkner
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA.
| | - Emily Lluch
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Simone Kennard
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Galina Antonova
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA, 30912, USA.,Department of Cardiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
42
|
Goto K, Kitazono T. Endothelium-dependent hyperpolarization (EDH) in diet-induced obesity. ENDOCRINE AND METABOLIC SCIENCE 2020. [DOI: 10.1016/j.endmts.2020.100062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
43
|
Faulkner JL, Belin de Chantemèle EJ. Female Sex, a Major Risk Factor for Salt-Sensitive Hypertension. Curr Hypertens Rep 2020; 22:99. [PMID: 33089375 PMCID: PMC7675065 DOI: 10.1007/s11906-020-01113-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW High dietary salt is a significant contributor to essential hypertension in clinical populations. However, although clinical studies indicate a higher prevalence of salt sensitivity in women over men, knowledge of salt-sensitive mechanisms is largely restricted to males, and female-specific mechanisms are presently being elucidated. RECENT FINDINGS Male-specific mechanisms of salt-sensitive hypertension are well published and predominantly appear to involve dysfunctional renal physiology. However, emerging novel evidence indicates that aldosterone production is sex-specifically heightened in salt-sensitive hypertensive women and female rodent models, which may be regulated by intra-adrenal renin-angiotensin system activation and sex hormone receptors. In addition, new evidence that young females endogenously express higher levels of endothelial mineralocorticoid receptors (MRs) and that endothelial MR is a crucial mediator of endothelial dysfunction in females indicates that the aldosterone-endothelial MR activation pathway is a novel mediator of salt-sensitive hypertension. Heightened aldosterone levels and endothelial MR expression provide a 2-fold sex-specific mechanism that may underlie the pathology of salt-sensitive hypertension in women. This hypothesis indicates that MR antagonists may be a preferential treatment for premenopausal women diagnosed with salt-sensitive hypertension.
Collapse
Affiliation(s)
- Jessica L Faulkner
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA.
- Department of Medicine (Cardiology), Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd., Augusta, GA, 30912, USA.
| |
Collapse
|
44
|
Shabbir A, Rathod KS, Khambata RS, Ahluwalia A. Sex Differences in the Inflammatory Response: Pharmacological Opportunities for Therapeutics for Coronary Artery Disease. Annu Rev Pharmacol Toxicol 2020; 61:333-359. [PMID: 33035428 DOI: 10.1146/annurev-pharmtox-010919-023229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Coordinated molecular responses are key to effective initiation and resolution of both acute and chronic inflammation. Vascular inflammation plays an important role in initiating and perpetuating atherosclerotic disease, specifically at the site of plaque and subsequent fibrous cap rupture. Both men and women succumb to this disease process, and although management strategies have focused on revascularization and pharmacological therapies in the acute situation to reverse vessel closure and prevent thrombogenesis, data now suggest that regulation of host inflammation may improve both morbidity and mortality, thus supporting the notion that prevention is better than cure. There is a clear sex difference in the incidence of vascular disease, and data confirm biological differences in inflammatory initiation and resolution between men and women. This article reviews contemporary opinions describing the sex difference in the initiation and resolution of inflammatory responses, with a view to explore potential targets for pharmacological intervention.
Collapse
Affiliation(s)
- Asad Shabbir
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Krishnaraj Sinhji Rathod
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Rayomand Syrus Khambata
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Amrita Ahluwalia
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| |
Collapse
|
45
|
Bar A, Targosz-Korecka M, Suraj J, Proniewski B, Jasztal A, Marczyk B, Sternak M, Przybyło M, Kurpińska A, Walczak M, Kostogrys RB, Szymonski M, Chlopicki S. Degradation of Glycocalyx and Multiple Manifestations of Endothelial Dysfunction Coincide in the Early Phase of Endothelial Dysfunction Before Atherosclerotic Plaque Development in Apolipoprotein E/Low-Density Lipoprotein Receptor-Deficient Mice. J Am Heart Assoc 2020; 8:e011171. [PMID: 30866689 PMCID: PMC6475045 DOI: 10.1161/jaha.118.011171] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background The impairment of endothelium‐dependent vasodilation, increased endothelial permeability, and glycocalyx degradation are all important pathophysiological components of endothelial dysfunction. However, it is still not clear whether in atherosclerosis, glycocalyx injury precedes other features of endothelial dysfunction or these events coincide. Methods and Results Herein, we demonstrate that in 4‐ to 8‐week‐old apolipoprotein E/low‐density lipoprotein receptor‐deficient mice, at the stage before development of atherosclerotic plaques, impaired acetylcholine‐induced vasodilation, reduced NO production in aorta, and increased endothelial permeability were all observed; however, flow‐mediated dilation in the femoral artery was fully preserved. In 4‐week‐old mice, glycocalyx coverage was reduced and endothelial stiffness was increased, whereas glycocalyx length was significantly decreased at 8 weeks of age. Early changes in endothelial function were also featured by increased plasma concentration of biomarkers of glycocalyx disruption (endocan), biomarkers of endothelial inflammation (soluble vascular cell adhesion molecule 1), increased vascular permeability (angiopoietin 2), and alterations in hemostasis (tissue plasminogen activator and plasminogen activator inhibitor 1). In 28‐week‐old mice, at the stage of advanced atherosclerotic plaque development, impaired NO production and nearly all other features of endothelial dysfunction were changed to a similar extent, compared with the preatherosclerotic plaque phase. The exceptions were the occurrence of acetylcholine‐induced vasoconstriction in the aorta and brachiocephalic artery, impaired flow‐mediated vasodilation in the femoral artery, and further reduction of glycocalyx length and coverage with a concomitant further increase in endothelial permeability. Conclusions In conclusion, even at the early stage before the development of atherosclerotic plaques, endothelial dysfunction is a complex multifactorial response that has not been previously appreciated.
Collapse
Affiliation(s)
- Anna Bar
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland.,3 Jagiellonian University Medical College Faculty of Medicine Chair of Pharmacology Krakow Poland
| | - Marta Targosz-Korecka
- 2 Center for Nanometer-Scale Science and Advanced Materials NANOSAM Faculty of Physics, Astronomy and Applied Computer Science Krakow Poland
| | - Joanna Suraj
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland.,4 Faculty of Pharmacy Chair and Department of Toxicology Krakow Poland
| | - Bartosz Proniewski
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland
| | - Agnieszka Jasztal
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland
| | - Brygida Marczyk
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland.,3 Jagiellonian University Medical College Faculty of Medicine Chair of Pharmacology Krakow Poland
| | - Magdalena Sternak
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland
| | - Magdalena Przybyło
- 5 Wroclaw University of Science and Technology Department of Biomedical Engineering Wroclaw Poland
| | - Anna Kurpińska
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland
| | - Maria Walczak
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland.,4 Faculty of Pharmacy Chair and Department of Toxicology Krakow Poland
| | - Renata B Kostogrys
- 6 University of Agriculture H. Kollataja in Cracow Department of Human Nutrition Faculty of Food Technology Krakow Poland
| | - Marek Szymonski
- 2 Center for Nanometer-Scale Science and Advanced Materials NANOSAM Faculty of Physics, Astronomy and Applied Computer Science Krakow Poland
| | - Stefan Chlopicki
- 1 Jagiellonian University Jagiellonian Centre for Experimental Therapeutics Krakow Poland.,3 Jagiellonian University Medical College Faculty of Medicine Chair of Pharmacology Krakow Poland
| |
Collapse
|
46
|
Manrique-Acevedo C, Chinnakotla B, Padilla J, Martinez-Lemus LA, Gozal D. Obesity and cardiovascular disease in women. Int J Obes (Lond) 2020; 44:1210-1226. [PMID: 32066824 PMCID: PMC7478041 DOI: 10.1038/s41366-020-0548-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/20/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022]
Abstract
As the prevalence of obesity continues to grow worldwide, the health and financial burden of obesity-related comorbidities grows too. Cardiovascular disease (CVD) is clearly associated with increased adiposity. Importantly, women are at higher risk of CVD when obese and insulin resistant, in particular at higher risk of developing heart failure with preserved ejection fraction and ischemic heart disease. Increased aldosterone and mineralocorticoid receptor activation, aberrant estrogenic signaling and elevated levels of androgens are among some of the proposed mechanisms explaining the heightened CVD risk. In addition to traditional cardiovascular risk factors, understanding nontraditional risk factors specific to women, like excess weight gain during pregnancy, preeclampsia, gestational diabetes, and menopause are central to designing personalized interventions aimed to curb the epidemic of CVD. In the present review, we examine the available evidence supporting a differential cardiovascular impact of increased adiposity in women compared with men and the proposed pathophysiological mechanisms behind these differences. We also discuss women-specific cardiovascular risk factors associated with obesity and insulin resistance.
Collapse
Affiliation(s)
- Camila Manrique-Acevedo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, USA
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Bhavana Chinnakotla
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - David Gozal
- Department of Child Health, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
47
|
Edwards JM, Roy S, Tomcho JC, Schreckenberger ZJ, Chakraborty S, Bearss NR, Saha P, McCarthy CG, Vijay-Kumar M, Joe B, Wenceslau CF. Microbiota are critical for vascular physiology: Germ-free status weakens contractility and induces sex-specific vascular remodeling in mice. Vascul Pharmacol 2020; 125-126:106633. [PMID: 31843471 PMCID: PMC7036036 DOI: 10.1016/j.vph.2019.106633] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/25/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
Abstract
Commensal microbiota within a holobiont contribute to the overall health of the host via mutualistic symbiosis. Disturbances in such symbiosis is prominently correlated with a variety of diseases affecting the modern society of humans including cardiovascular diseases, which are the number one contributors to human mortality. Given that a hallmark of all cardiovascular diseases is changes in vascular function, we hypothesized that depleting microbiota from a holobiont would induce vascular dysfunction. To test this hypothesis, young mice of both sexes raised in germ-free conditions were examined vascular contractility and structure. Here we observed that male and female germ-free mice presented a decrease in contraction of resistance arteries. These changes were more pronounced in germ-free males than in germ-free females mice. Furthermore, there was a distinct change in vascular remodeling between males and females germ-free mice. Resistance arteries from male germ-free mice demonstrated increased vascular stiffness, as shown by the leftward shift in the stress-strain curve and inward hypotrophic remodeling, a characteristic of chronic reduction in blood flow. On the other hand, resistance arteries from germ-free female mice were similar in the stress-strain curves to that of conventionally raised mice, but were distinctly different and showed outward hypertrophic remodeling, a characteristic seen in aging. Interestingly, we observed that reactive oxygen species (ROS) generation from bone marrow derived neutrophils is blunted in female germ-free mice, but it is exacerbated in male germ-free mice. In conclusion, these observations indicate that commensal microbiota of a holobiont are central to maintain proper vascular function and structure homeostasis, especially in males.
Collapse
Affiliation(s)
- Jonnelle M Edwards
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Shaunak Roy
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Jeremy C Tomcho
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Zachary J Schreckenberger
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Saroj Chakraborty
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Nicole R Bearss
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Piu Saha
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Cameron G McCarthy
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Matam Vijay-Kumar
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Bina Joe
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA
| | - Camilla F Wenceslau
- Microbiome Consortium, Center for Hypertension and Precision Medicine, Department of Physiology and Pharmacology, University of Toledo College of Medicine & Life Sciences, Toledo, OH, USA.
| |
Collapse
|
48
|
Norton CE, Jacobsen NL, Sinkler SY, Manrique-Acevedo C, Segal SS. Female sex and Western-style diet protect mouse resistance arteries during acute oxidative stress. Am J Physiol Cell Physiol 2019; 318:C627-C639. [PMID: 31891519 DOI: 10.1152/ajpcell.00342.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A Western-style diet (WD; high in fat and carbohydrates) increases vascular oxidative stress. We hypothesized that vascular cells adapt to a WD by developing resilience to oxidative stress. Male and female C57BL/6J mice (4 wk of age) were fed a control diet (CD) or a WD for 16-20 wk. Superior epigastric arteries (SEAs; diameter, ~125 µm) were isolated and pressurized for study. Basal reactive oxygen species production was greatest in SEAs from males fed the WD. During exposure to H2O2 (200 μM, 50 min), propidium iodide staining identified nuclei of disrupted endothelial cells (ECs) and smooth muscle cells (SMCs). For mice fed the CD, death of SMCs (21%) and ECs (6%) was greater (P < 0.05) in SEAs from males than females (9% and 2%, respectively). WD consumption attenuated cell death most effectively in SEAs from males. With no difference at rest, H2O2 increased intracellular Ca2+ concentration ([Ca2+]i) to the greatest extent in SEAs from males, as shown by fura 2 fluorescence. Selective disruption of the endothelium (luminal air bubble) increased [Ca2+]i and SMC death during H2O2 exposure irrespective of sex; the WD reduced both responses most effectively in males. Nonselective transient receptor potential (TRP) channel inhibition (ruthenium red, 5 μM) attenuated the rise of [Ca2+]i, as did selective inhibition of TRP vanilloid type 4 (TRPV4) channels (HC-067047, 1 μM), which also attenuated cell death. In contrast, inhibition of voltage-gated Ca2+ channels (diltiazem, 50 μM) was without effect. Thus, for resistance arteries during acute oxidative stress: 1) ECs are more resilient than (and can protect) SMCs, 2) vessels from females are inherently more resilient than those from males, and 3) a WD increases vascular resilience by diminishing TRPV4 channel-dependent Ca2+ entry.
Collapse
Affiliation(s)
- Charles E Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Nicole L Jacobsen
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Shenghua Y Sinkler
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Department of Medicine, University of Missouri, Columbia, Missouri.,Research Services, Harry S Truman Memorial Veterans Hospital, Columbia, Missouri.,Dalton Cardiovascular Research Center, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, Columbia, Missouri
| |
Collapse
|
49
|
Padilla J, Woodford ML, Lastra-Gonzalez G, Martinez-Diaz V, Fujie S, Yang Y, Lising AMC, Ramirez-Perez FI, Aroor AR, Morales-Quinones M, Ghiarone T, Whaley-Connell A, Martinez-Lemus LA, Hill MA, Manrique-Acevedo C. Sexual Dimorphism in Obesity-Associated Endothelial ENaC Activity and Stiffening in Mice. Endocrinology 2019; 160:2918-2928. [PMID: 31617909 PMCID: PMC6853665 DOI: 10.1210/en.2019-00483] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 10/10/2019] [Indexed: 02/08/2023]
Abstract
Obesity and insulin resistance stiffen the vasculature, with females appearing to be more adversely affected. As augmented arterial stiffness is an independent predictor of cardiovascular disease (CVD), the increased predisposition of women with obesity and insulin resistance to arterial stiffening may explain their heightened risk for CVD. However, the cellular mechanisms by which females are more vulnerable to arterial stiffening associated with obesity and insulin resistance remain largely unknown. In this study, we provide evidence that female mice are more susceptible to Western diet-induced endothelial cell stiffening compared with age-matched males. Mechanistically, we show that the increased stiffening of the vascular intima in Western diet-fed female mice is accompanied by enhanced epithelial sodium channel (ENaC) activity in endothelial cells (EnNaC). Our data further indicate that: (i) estrogen signaling through estrogen receptor α (ERα) increases EnNaC activity to a larger extent in females compared with males, (ii) estrogen-induced activation of EnNaC is mediated by the serum/glucocorticoid inducible kinase 1 (SGK-1), and (iii) estrogen signaling stiffens endothelial cells when nitric oxide is lacking and this stiffening effect can be reduced with amiloride, an ENaC inhibitor. In aggregate, we demonstrate a sexual dimorphism in obesity-associated endothelial stiffening, whereby females are more vulnerable than males. In females, endothelial stiffening with obesity may be attributed to estrogen signaling through the ERα-SGK-1-EnNaC axis, thus establishing a putative therapeutic target for female obesity-related vascular stiffening.
Collapse
Affiliation(s)
- Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Makenzie L Woodford
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Guido Lastra-Gonzalez
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
| | - Vanesa Martinez-Diaz
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
| | - Shumpei Fujie
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Faculty of Sport and Health Sciences, University of Tsukuba, Ibaraki, Japan
- Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Alexandre M C Lising
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Francisco I Ramirez-Perez
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biological Engineering, University of Missouri, Columbia, Missouri
| | - Annayya R Aroor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
| | | | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Adam Whaley-Connell
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
- Division of Nephrology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Biological Engineering, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, Missouri
- Research Service, Harry S. Truman Memorial Veterans’ Hospital, Columbia, Missouri
- Correspondence: Camila Manrique-Acevedo, MD, Department of Medicine, University of Missouri, D109 Diabetes Center UHC, One Hospital Drive, Columbia, Missouri 65212. E-mail:
| |
Collapse
|
50
|
Moss ME, Carvajal B, Jaffe IZ. The endothelial mineralocorticoid receptor: Contributions to sex differences in cardiovascular disease. Pharmacol Ther 2019; 203:107387. [PMID: 31271793 PMCID: PMC6848769 DOI: 10.1016/j.pharmthera.2019.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/26/2019] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease remains the leading cause of death for both men and women. The observation that premenopausal women are protected from cardiovascular disease relative to age-matched men, and that this protection is lost with menopause, has led to extensive study of the role of sex steroid hormones in the pathogenesis of cardiovascular disease. However, the molecular basis for sex differences in cardiovascular disease is still not fully understood, limiting the ability to tailor therapies to male and female patients. Therefore, there is a growing need to investigate molecular pathways outside of traditional sex hormone signaling to fully understand sex differences in cardiovascular disease. Emerging evidence points to the mineralocorticoid receptor (MR), a steroid hormone receptor activated by the adrenal hormone aldosterone, as one such mediator of cardiovascular disease risk, potentially serving as a sex-dependent link between cardiovascular risk factors and disease. Enhanced activation of the MR by aldosterone is associated with increased risk of cardiovascular disease. Emerging evidence implicates the MR specifically within the endothelial cells lining the blood vessels in mediating some of the sex differences observed in cardiovascular pathology. This review summarizes the available clinical and preclinical literature concerning the role of the MR in the pathophysiology of endothelial dysfunction, hypertension, atherosclerosis, and heart failure, with a special emphasis on sex differences in the role of endothelial-specific MR in these pathologies. The available data regarding the molecular mechanisms by which endothelial-specific MR may contribute to sex differences in cardiovascular disease is also summarized. A paradigm emerges from synthesis of the literature in which endothelial-specific MR regulates vascular function in a sex-dependent manner in response to cardiovascular risk factors to contribute to disease. Limitations in this field include the relative paucity of women in clinical trials and, until recently, the nearly exclusive use of male animals in preclinical investigations. Enhanced understanding of the sex-specific roles of endothelial MR could lead to novel mechanistic insights underlying sex differences in cardiovascular disease incidence and outcomes and could identify additional therapeutic targets to effectively treat cardiovascular disease in men and women.
Collapse
Affiliation(s)
- M Elizabeth Moss
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America
| | - Brigett Carvajal
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America.
| |
Collapse
|