1
|
Dimitriadis K, Theofilis P, Koutsopoulos G, Pyrpyris N, Beneki E, Tatakis F, Tsioufis P, Chrysohoou C, Fragkoulis C, Tsioufis K. The role of coronary microcirculation in heart failure with preserved ejection fraction: An unceasing odyssey. Heart Fail Rev 2025; 30:75-88. [PMID: 39358622 DOI: 10.1007/s10741-024-10445-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents an entity with complex pathophysiologic pathways, among which coronary microvascular dysfunction (CMD) is believed to be an important orchestrator. Research in the field of CMD has highlighted impaired vasoreactivity, capillary rarefaction, and inflammation as potential mediators of its development. CMD can be diagnosed via several noninvasive methods including transthoracic echocardiography, cardiac magnetic resonance, and positron emission tomography. Moreover, invasive methods such as coronary flow reserve and index of microcirculatory resistance are commonly employed in the assessment of CMD. As far as the association between CMD and HFpEF is concerned, numerous studies have highlighted the coexistence of CMD in the majority of HFpEF patients. Additionally, patients affected by both conditions may be facing an adverse prognosis. Finally, there is limited evidence suggesting a beneficial effect of renin-angiotensin-aldosterone system blockers, ranolazine, and sodium-glucose cotransporter-2 inhibitors in CMD, with further evidence being awaited regarding the impact of other pharmacotherapies such as anti-inflammatory agents.
Collapse
Affiliation(s)
- Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece.
| | - Panagiotis Theofilis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Georgios Koutsopoulos
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Eirini Beneki
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Fotis Tatakis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Panagiotis Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Christina Chrysohoou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Christos Fragkoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, Vasilissis Sofias 114, 11528, Athens, PO, Greece
| |
Collapse
|
2
|
Mooradian AD. Cardiomodulatory Effects of Cardiometabolic and Antihyperglycemic Medications: The Roles of Oxidative and Endoplasmic Reticulum Stress. Am J Cardiovasc Drugs 2025; 25:37-46. [PMID: 39392561 DOI: 10.1007/s40256-024-00685-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/12/2024]
Abstract
Uncontrolled hyperglycemia in people with diabetes is an established risk of premature cardiovascular disease. Repeated hypoglycemic events are also associated with increased cardiovascular mortality. Both hyperglycemia and hypoglycemia induce cellular stress, notably endoplasmic reticulum (ER) stress, a known promoter of cardiovascular disease. Contemporary anti-hyperglycemic drugs such as glucagon-like peptide 1 (GLP-1) receptor agonists and sodium-glucose cotransporter 2 (SGLT-2) inhibitors simultaneously inhibit oxidative stress and ER stress in human coronary artery endothelial cells. Similarly, other known cardioprotective drugs, such as statins and inhibitors of the renin-angiotensin-aldosterone system (RAAS) share a common pleiotropic effect of reducing cellular stress. Antioxidants reduce oxidative stress but may aggravate ER stress. This dichotomy of antioxidant effects may underline the unfavorable outcomes of clinical trials with antioxidant vitamin use. The aim of this review is to highlight the potential role of cellular stress reduction in cardioprotective effects of contemporary diabetes drugs. Future clinical trials are needed to test the hypothesis that cellular stress is the fundamental culprit in cardiovascular disease.
Collapse
Affiliation(s)
- Arshag D Mooradian
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Florida Jacksonville College of Medicine, 653-1 West 8th Street, Jacksonville, FL, 32209, USA.
| |
Collapse
|
3
|
Tryon D, Corban MT, Alkhouli M, Prasad A, Raphael CE, Rihal CS, Reeder GS, Lewis B, Albers D, Gulati R, Lerman A. Coronary Sinus Reducer Improves Angina, Quality of Life, and Coronary Flow Reserve in Microvascular Dysfunction. JACC Cardiovasc Interv 2024; 17:2893-2904. [PMID: 39520443 DOI: 10.1016/j.jcin.2024.09.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Coronary microvascular dysfunction (CMD) is a common cause of angina with no obstructive coronary artery disease (ANOCA), and effective treatment options are limited. OBJECTIVES This study aims to assess the safety and efficacy of the coronary sinus (CS) Reducer (Neovasc, Inc/Shockwave Medical) for treatment of angina in patients with CMD. METHODS This Phase II trial enrolled 30 patients with ANOCA, invasively diagnosed CMD, and Canadian Cardiovascular Society (CCS) class 3 to 4 angina despite medical therapy. CMD was defined by coronary flow reserve (CFR) ≤2.5 and/or ≤50% increase in coronary blood flow (CBF) in response to intracoronary infusion of acetylcholine. Invasive coronary microvascular function testing was performed before and at 120 days postimplantation. The primary endpoint was change in microvascular function at 120 days. Secondary endpoints were changes in CCS angina class and Seattle Angina Questionnaire (SAQ) scores. RESULTS Mean age was 54.8 ± 11.0 years; 67% (20/30) were women. In patients with low baseline CFR (endothelium-independent CMD), CFR increased significantly from 2.1 (1.95-2.30) to 2.7 (2.45-2.95) (n = 19; P = 0.0011). Patients with abnormal CBF response to acetylcholine at baseline (endothelium-dependent CMD) had an increase in CBF response to acetylcholine: -11.0% (-20.15% to 5.85%) to 11.5% (-4.82% to 39.29%) (n = 11; P = 0.042). There was a significant improvement in CCS angina class from 4.0 (3.25-4.0) to 2.0 (2.0-3.0) (P < 0.001) and increase in each domain of the SAQ questionnaire (P < 0.006 for all). CONCLUSIONS This study demonstrates that the CS Reducer is associated with significant improvement in angina, quality of life, and coronary microvascular function in patients with CMD and may emerge as a novel therapy for patients with ANOCA.
Collapse
Affiliation(s)
| | - Michel T Corban
- Mayo Clinic, Rochester, Minnesota, USA; Sarver Heart Center, University of Arizona College of Medicine Tucson, Arizona, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Badran M, Khalyfa A, Bailey CA, Gozal D, Bender SB. Mineralocorticoid receptor antagonism prevents coronary microvascular dysfunction in intermittent hypoxia independent of blood pressure. Sleep 2024:zsae296. [PMID: 39693220 DOI: 10.1093/sleep/zsae296] [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: 07/29/2024] [Indexed: 12/20/2024] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea (OSA), characterized by intermittent hypoxia (IH), and is associated with increased cardiovascular mortality that may not be reduced by standard therapies. Inappropriate activation of the renin-angiotensin-aldosterone system occurs in IH, and mineralocorticoid receptor (MR) blockade has been shown to improve vascular outcomes in cardiovascular disease. Thus, we hypothesized that MR inhibition prevents coronary and renal vascular dysfunction in mice exposed to chronic IH. METHODS Human and mouse coronary vascular cells and male C57BL/6J mice were exposed to IH or room air (RA) for 12 hours/day for 3 days (in vitro) and 6 weeks with or without treatments with spironolactone (SPL) or hydrochlorothiazide (HTZ). RESULTS In vitro studies demonstrated that IH increased MR gene expression in human and mouse coronary artery endothelial and smooth muscle cells. Exposure to IH in mice increased blood pressure, reduced coronary flow velocity reserve (CFVR), and attenuated endothelium-dependent dilation and enhanced vasoconstrictor responsiveness in coronary, but not renal arteries. Importantly, SPL treatment prevented altered coronary vascular function independent of blood pressure as normalization of BP with HTZ did not improve CFVR or coronary vasomotor function. CONCLUSIONS These data demonstrate that chronic IH, which mimics the hypoxia-reoxygenation cycles of moderate-to-severe OSA, increases coronary vascular MR expression in vitro. It also selectively promotes coronary vascular dysfunction in mice. Importantly, this dysfunction is sensitive to MR antagonism by SPL, independent of blood pressure. These findings suggest that MR blockade could serve as an adjuvant therapy to improve long-term cardiovascular outcomes in patients with OSA.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Pediatrics, University of Missouri; Columbia, MO, USA
- Department of Medical Physiology and Pharmacology, University of Missouri; Columbia, MO, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Marshall University; Huntington, WV, USA
| | - Chastidy A Bailey
- Department of Biomedical Sciences, University of Missouri; Columbia, MO, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital; Columbia, MO, United States
| | - David Gozal
- Department of Pediatrics, University of Missouri; Columbia, MO, USA
- Office of the Dean, Joan C. Edwards School of Medicine, Marshall University; Huntington, WV, USA
| | - Shawn B Bender
- Department of Biomedical Sciences, University of Missouri; Columbia, MO, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital; Columbia, MO, United States
- Dalton Cardiovascular Research Center, University of Missouri; Columbia, MO, United States
| |
Collapse
|
5
|
Binu AJ, Kapoor N. Understanding Diabetic Cardiomyopathy: Insulin Resistance and Beyond. Heart Int 2024; 18:7-13. [PMID: 39885933 PMCID: PMC11781369 DOI: 10.17925/hi.2024.18.2.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/16/2024] [Indexed: 02/01/2025] Open
Abstract
Background: Diabetic cardiomyopathy (DC) is a syndrome of heart failure occurring in patients with diabetes mellitus (DM), independent of other risk factors. It is a relatively underdiagnosed condition with a prolonged subclinical phase. There is an abundance of studies put forward to explain the underlying pathogenic mechanisms observed in this condition. This review aims to summarize the evidence available in contemporary medical literature with regard to the molecular mechanisms, abnormalities in signalling and metabolism and structural and functional abnormalities manifesting as DC. Methods: We conducted a literature search using the terms 'diabetic cardiomyopathy', 'heart failure AND Diabetes mellitus', 'Cardiomyopathy AND Diabetes mellitus'. We searched the reference lists of included studies and relevant systematic reviews. Results: In this review, we elucidate all the mechanisms that have been postulated to have a role in the pathogenesis of DC, in addition to insulin resistance, such as inflammation, renin-angiotensin-aldosterone system activation and deranged protein homeostasis. Conclusions: DC is an underrecognized cardiac complication of DM. A comprehensive knowledge of all the pathways and mediators will aid in the development of diagnostic and prognostic markers, screening protocols and novel management strategies.
Collapse
Affiliation(s)
- Aditya John Binu
- Department of Cardiology, Christian Medical College & Hospital, Vellore, Tamil Nadu, India
| | - Nitin Kapoor
- Department of Endocrinology, Diabetes and Metabolism, Christian Medical College & Hospital, Vellore, Tamil Nadu, India
- Non-Communicable Disease Unit, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Adji AS, Widjaja JS, de Liyis BG. Effectiveness and safety of mineralocorticoid receptor antagonists in heart failure patients with and without diabetes: a systematic review and meta-analysis. Egypt Heart J 2024; 76:150. [PMID: 39541086 PMCID: PMC11564587 DOI: 10.1186/s43044-024-00580-5] [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: 06/15/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Mineralocorticoid receptor antagonists (MRAs) have been shown to improve outcomes in various populations of heart failure (HF) patients. However, the impact of concomitant diseases, such as diabetes mellitus (DM), on these outcomes remains unclear. This meta-analysis aimed to evaluate the efficacy and safety of MRAs in heart failure patients with and without diabetes mellitus. METHODS A systematic search was conducted on PubMed, Scopus, and Google Scholar databases up to April 30, 2024. Data analysis was performed using a random-effects model to account for variability across studies, and statistical analysis was carried out using Review Manager 5.4. Efficacy and safety parameters were evaluated in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis guidelines. RESULTS The meta-analysis included a total of 21,832 subjects from ten studies. The pooled results demonstrated that MRAs, compared to placebo, significantly reduced all-cause mortality in HF patients with and without DM (RR: 0.85; 95%CI 0.75-0.96; p = 0.009). A similar effect was observed in HF patients without DM (RR: 0.83; 95%CI 0.71-0.97; p = 0.02), while no significant effect was detected in the DM subgroup (RR: 0.87; 95%CI 0.69-1.11; p = 0.27). Both treatments had comparable effects on cardiovascular mortality in HF patients with and without DM (RR: 0.88; 95%CI 0.82-0.94; p = 0.0002), in HF patients with DM (RR: 0.90; 95%CI 0.81-1.01; p = 0.08), and in the non-DM subgroup (RR: 0.86; 95%CI 0.79-0.94; p = 0.0009). MRAs significantly reduced the risk of cardiovascular mortality in HF patients with and without DM (RR: 0.82; 95%CI 0.72-0.94; p = 0.005) and in HF patients with DM (RR: 0.79; 95%CI 0.63-0.98; p = 0.03), but no significant effect was observed in the non-DM subgroup (RR: 0.85; 95%CI 0.69-1.05; p = 0.13). Furthermore, compared to placebo, MRAs were associated with an increased risk of hyperkalemia (> 5.5 mEq/L) in HF patients with and without DM (RR: 1.63; 95%CI 1.18-2.24; p = 0.003), particularly in HF patients with DM (RR: 1.44; 95%CI 0.97-2.13; p = 0.07) and in the non-DM subgroup (RR: 1.87; 95%CI 1.34-2.61; p = 0.0002). CONCLUSION MRAs are effective in reducing all-cause mortality, cardiovascular death, and cardiovascular mortality in heart failure patients. However, the use of MRAs is associated with an increased risk of hyperkalemia, necessitating careful monitoring, particularly in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Arga Setyo Adji
- Faculty of Medicine, Hang Tuah University, Ahmad Yani Street no.1, Wonokromo, Surabaya, East Java, 60244, Indonesia.
| | - Jordan Steven Widjaja
- Faculty of Medicine, Hang Tuah University, Ahmad Yani Street no.1, Wonokromo, Surabaya, East Java, 60244, Indonesia
| | | |
Collapse
|
7
|
Taguchi K, Kondo H, Matsumoto T, Kobayashi T. Effect of esaxerenone on the onset of aortic endothelial dysfunction and circulating microparticles in type 1 diabetic male mice. Sci Rep 2024; 14:26266. [PMID: 39487333 PMCID: PMC11530539 DOI: 10.1038/s41598-024-78321-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024] Open
Abstract
Endothelial dysfunction exacerbates hypertension and other vascular complications in diabetes mellitus (DM). Circulating microparticles (MPs) and extracellular vesicles released in patients with DM have emerged as novel regulators of endothelial dysfunction. The obstruction of mineralocorticoid receptors (MRs) is a potential therapeutic approach to reduce cardiovascular complications. Their impact on the obstruction of MRs on circulating MPs and endothelial dysfunction in DM remains unclear. DM was induced in mice through a single intravenous dose of streptozotocin (STZ; 200 mg/kg). Esaxerenone (ESAX; 3 mg/kg/day), a MR blocker was administered via diet for 8 weeks. In this study, the aortas of the DM group showed the endothelial dysfunction and the administration of ESAX ameliorated the endothelial-dependent responses. Moreover, ESAX influences the impaired endothelial-dependent responses of DM-derived MPs. Interestingly, MP levels increased in DM whereas decreased after ESAX administration. In the aorta, the DM-derived MPs increased the expression of intercellular adhesion molecule-1 (ICAM-1). ESAX inhibited the adhesion of DM-derived MPs. Moreover, the ICAM-1 inhibitor A205804 shows similar effects as ESAX. These results indicate that the release and adhesion properties of MPs can be partially obstructed by ESAX via the ICAM-1 signaling pathway, which clarifies the other functions beyond the anti-hypertensive effects of ESAX.
Collapse
MESH Headings
- Animals
- Cell-Derived Microparticles/metabolism
- Cell-Derived Microparticles/drug effects
- Male
- Mice
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Aorta/drug effects
- Aorta/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/complications
- Pyrroles/pharmacology
- Sulfones/pharmacology
- Mice, Inbred C57BL
- Mineralocorticoid Receptor Antagonists/pharmacology
- Receptors, Mineralocorticoid/metabolism
Collapse
Affiliation(s)
- Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Hiroyuki Kondo
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, 1714-1 Yoshino-cho, Nobeoka, 882-8508, Miyazaki, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan.
| |
Collapse
|
8
|
Srinivasa S, Walpert AR, Huck D, Thomas TS, Dunderdale CN, Lee H, Dicarli MF, Adler GK, Grinspoon SK. Coronary Microvascular Dysfunction Is Present Among Well-Treated Asymptomatic Persons With HIV and Similar to Those With Diabetes. Open Forum Infect Dis 2024; 11:ofae234. [PMID: 38813261 PMCID: PMC11134457 DOI: 10.1093/ofid/ofae234] [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: 03/27/2024] [Accepted: 04/24/2024] [Indexed: 05/31/2024] Open
Abstract
Background Coronary microvascular dysfunction (CMD) could be a potential underlying mechanism for myocardial disease in HIV. Methods Comparisons of coronary flow reserve corrected for heart rate-blood pressure product (CFRCOR) were made among people with HIV (PWH) with no known history of cardiovascular disease (CVD) or diabetes mellitus, persons without HIV (PWOH), and persons with diabetes (PWDM) and no known history of CVD or HIV. Results PWH (n = 39, 74% male, age 55 [7] years, body mass index [BMI] 32.3 (26.8-34.9) kg/m2, duration of antiretroviral therapy 13 [5] years, CD4+ count 754 [598-961] cells/μL) were similar to PWOH (n = 69, 74% male, age 55 [8] years, BMI 32.2[25.6-36.5] kg/m2) and PWDM (n = 63, 63% male, age 55 [8] years, BMI 31.5 [28.6-35.6] kg/m2). CFRCOR was different among groups: PWOH 2.76 (2.37-3.36), PWH 2.47 (1.92-2.93), and PWDM 2.31 (1.98-2.84); overall P = .003. CFRCOR was reduced comparing PWH to PWOH (P = .04) and PWDM to PWOH (P = .007) but did not differ when comparing PWH to PWDM (P = .98). A total 31% of PWH had CFRCOR < 2.0, a critical cutoff for CMD, compared to 14% of PWOH and 27% with PWDM. A total 40% of women with HIV had a CFRCOR < 2.0 compared to 6% of women without HIV (P = .02). Conclusions Subclinical CMD is present among chronically infected and well-treated, asymptomatic PWH who are immunologically controlled. This study demonstrates CFR is reduced in PWH compared to PWOH and comparable to PWDM, further highlighting that well-treated HIV infection is a CVD-risk enhancing factor for CMD similar to diabetes. Clinical Trials Registration: NCT02740179.
Collapse
Affiliation(s)
- Suman Srinivasa
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Allie R Walpert
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Huck
- Division of Nuclear Medicine and Molecular Imaging, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Teressa S Thomas
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Carolyn N Dunderdale
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marcelo F Dicarli
- Division of Nuclear Medicine and Molecular Imaging, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gail K Adler
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Brown JM. Adverse Effects of Aldosterone: Beyond Blood Pressure. J Am Heart Assoc 2024; 13:e030142. [PMID: 38497438 PMCID: PMC11179780 DOI: 10.1161/jaha.123.030142] [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] [Indexed: 03/19/2024]
Abstract
Aldosterone is a steroid hormone that primarily acts through activation of the mineralocorticoid receptor (MR), a nuclear receptor responsible for downstream genomic regulation. Classically, activation of the MR in the renal tubular epithelium is responsible for sodium retention and volume expansion, raising systemic blood pressure. However, activation of the MR across a wide distribution of tissue types has been implicated in multiple adverse consequences for cardiovascular, cerebrovascular, renal, and metabolic disease, independent of blood pressure alone. Primary aldosteronism, heart failure, and chronic kidney disease are states of excessive aldosterone production and MR activity where targeting MR activation has had clinical benefits out of proportion to blood pressure lowering. The growing list of established and emerging therapies that target aldosterone and MR activation may provide new opportunities to improve clinical outcomes and enhance cardiovascular and renal health.
Collapse
Affiliation(s)
- Jenifer M. Brown
- Division of Cardiovascular Medicine, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMAUSA
| |
Collapse
|
10
|
Finsen SH, Hansen MR, Hansen PBL, Mortensen SP. Eight weeks of treatment with mineralocorticoid receptor blockade does not alter vascular function in individuals with and without type 2 diabetes. Physiol Rep 2024; 12:e16010. [PMID: 38610066 PMCID: PMC11014871 DOI: 10.14814/phy2.16010] [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: 10/24/2023] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Aldosterone has been suggested to be involved in the microvascular complications observed in type 2 diabetes. We aimed to investigate the effect of mineralocorticoid receptor (MR) blockade on endothelial function in individuals with type 2 diabetes compared to healthy controls. We included 12 participants with type 2 diabetes and 14 controls. We measured leg hemodynamics at baseline and during femoral arterial infusion of acetylcholine and sodium nitroprusside before and 8 weeks into treatment with MR blockade (eplerenone). Acetylcholine infusion was repeated with concomitant n-acetylcysteine (antioxidant) infusion. No difference in leg blood flow or vascular conductance was detected before or after the treatment with MR blockade in both groups and there was no difference between groups. Infusion of n-acetylcysteine increased baseline blood flow and vascular conductance, but did not change the vascular response to acetylcholine before or after treatment with MR blockade. Skeletal muscle eNOS content was unaltered by MR blockade and no difference between groups was detected. In conclusion, we found no effect of MR blockade endothelial function in individuals with and without type 2 diabetes. As the individuals with type 2 diabetes did not have vascular dysfunction, these results might not apply to individuals with vascular dysfunction.
Collapse
Affiliation(s)
- Stine H. Finsen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of NephrologyOdense University HospitalOdenseDenmark
| | - Mie R. Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Pernille B. L. Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Stefan P. Mortensen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
11
|
Srinivasa S, Walpert AR, Thomas TS, Huck DM, Jerosch-Herold M, Islam S, Lu MT, Burdo TH, deFilippi CR, Dunderdale CN, Feldpausch M, Iyengar S, Shen G, Baak S, Torriani M, Robbins GK, Lee H, Kwong R, DiCarli M, Adler GK, Grinspoon SK. Randomized Placebo-Controlled Trial to Evaluate Effects of Eplerenone on Myocardial Perfusion and Function Among Persons With Human Immunodeficiency Virus (HIV)-Results From the MIRACLE HIV Study. Clin Infect Dis 2023; 77:1166-1175. [PMID: 37243345 PMCID: PMC10573745 DOI: 10.1093/cid/ciad310] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/01/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Increased renin angiotensin aldosterone system (RAAS) activity may contribute to excess cardiovascular disease in people with HIV (PWH). We investigated how RAAS blockade may improve myocardial perfusion, injury, and function among well-treated PWH. METHODS Forty PWH, on stable ART, without known heart disease were randomized to eplerenone 50 mg PO BID (n = 20) or identical placebo (n = 20) for 12 months. The primary endpoints were (1) myocardial perfusion assessed by coronary flow reserve (CFR) on cardiac PET or stress myocardial blood flow (sMBF) on cardiac MRI or (2) myocardial inflammation by extracellular mass index (ECMi) on cardiac MRI. RESULTS Beneficial effects on myocardial perfusion were seen for sMBF by cardiac MRI (mean [SD]: 0.09 [0.56] vs -0.53 [0.68] mL/min/g; P = .03) but not CFR by cardiac PET (0.01 [0.64] vs -0.07 [0.48]; P = .72, eplerenone vs placebo). Eplerenone improved parameters of myocardial function on cardiac MRI including left ventricular end diastolic volume (-13 [28] vs 10 [26] mL; P = .03) and global circumferential strain (GCS; median [interquartile range 25th-75th]: -1.3% [-2.9%-1.0%] vs 2.3% [-0.4%-4.1%]; P = .03), eplerenone versus placebo respectively. On cardiac MRI, improvement in sMBF related to improvement in global circumferential strain (ρ = -0.65, P = .057) among those treated with eplerenone. Selecting for those with impaired myocardial perfusion (CFR <2.5 and/or sMBF <1.8), there was a treatment effect of eplerenone versus placebo to improve CFR (0.28 [0.27] vs -0.05 [0.36]; P = .04). Eplerenone prevented a small increase in troponin (0.00 [-0.13-0.00] vs 0.00 [0.00-0.74] ng/L; P = .03) without effects on ECMi (0.9 [-2.3-4.3] vs -0.7 [-2.2--0.1] g/m2; P = .38). CD4+ T-cell count (127 [-38-286] vs -6 [-168-53] cells/μL; P = .02) increased in the eplerenone- versus placebo-treated groups. CONCLUSIONS RAAS blockade with eplerenone benefitted key indices and prevented worsening of myocardial perfusion, injury, and function among PWH with subclinical cardiac disease when compared with placebo. CLINICAL TRIALS REGISTRATION NCT02740179 (https://clinicaltrials.gov/ct2/show/NCT02740179?term=NCT02740179&draw=2&rank=1).
Collapse
Affiliation(s)
- Suman Srinivasa
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Allie R Walpert
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Teressa S Thomas
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel M Huck
- Division of Nuclear Medicine and Molecular Imaging, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Jerosch-Herold
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sabeeh Islam
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tricia H Burdo
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | | | - Carolyn N Dunderdale
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Meghan Feldpausch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sanjna Iyengar
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Grace Shen
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen Baak
- Department of Neuroscience, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Martin Torriani
- Division of Musculoskeletal Imaging and Intervention, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gregory K Robbins
- Division of Infectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Hang Lee
- Biostatistics Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Raymond Kwong
- Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marcelo DiCarli
- Division of Nuclear Medicine and Molecular Imaging, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gail K Adler
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Ahmed AI, Saad JM, Alahdab F, Han Y, Nayfeh M, Alfawara MS, Al-Rifai M, Al-Mallah M. Prognostic value of positron emission tomography derived myocardial flow reserve: A systematic review and meta-analysis. Atherosclerosis 2023; 382:117280. [PMID: 37742396 DOI: 10.1016/j.atherosclerosis.2023.117280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND AND AIMS Positron Emission Tomography (PET)-derived myocardial flow reserve (MFR) has been shown to have a role in the diagnosis and prognosis of patients with coronary artery disease (CAD). We performed a systematic review and meta-analysis to summarize the body of literature and synthesize the evidence on the prognostic role of PET-derived MFR in patients with known or suspected CAD. METHODS A comprehensive literature search of the Medline database from its inception to August 2023, in humans, in any language, was conducted for clinical studies examining the prognostic value of PET imaging in patients of any age, sex, and CAD status. Systematic screening and data extraction of the identified studies were followed by quantitative meta-analysis of PET-MFR's role in predicting adverse clinical events using random effect model. Studies were appraised using the modified Newcastle-Ottawa tool. RESULTS A total of 21 studies assessing the prognostic role of PET derived MFR in 46,815 patients with known and/or suspected CAD were included (mean (SD) age 66 (4) years, 48% women). The mean follow-up duration was 36 months (range 10-96). Cardiovascular risk factors were prevalent (73% hypertension, 35% diabetes and 67% dyslipidemia). The definition of the composite outcome varied between studies, with various combinations of mortality, non-fatal myocardial infarction, hospitalization, and coronary revascularization. Pooled impaired MFR was significantly associated with an increased risk of adverse outcomes (RR = 2.94, 95% CI 2.42-3.56, p < 0.001). Results were similar in a subgroup of patients with suspected CAD. CONCLUSIONS The available body of evidence shows that impaired PET-derived MFR measured using different tracers and PET systems is strongly associated with an increased risk of adverse cardiovascular events. Limitations of this review include observational nature of studies, marked heterogeneity in patient populations, inconsistency in thresholds to define abnormal MFR, and differing components for the composite outcome.
Collapse
Affiliation(s)
- Ahmed Ibrahim Ahmed
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA; Yale School of Medicine, New Haven, CT, USA
| | - Jean Michel Saad
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Fares Alahdab
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Yushui Han
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Malek Nayfeh
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | | | - Mahmoud Al-Rifai
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA
| | - Mouaz Al-Mallah
- Houston Methodist DeBakey Heart & Vascular Center, Houston, TX, USA.
| |
Collapse
|
13
|
Zhang Z, Li X, He J, Wang S, Wang J, Liu J, Wang Y. Molecular mechanisms of endothelial dysfunction in coronary microcirculation dysfunction. J Thromb Thrombolysis 2023; 56:388-397. [PMID: 37466848 DOI: 10.1007/s11239-023-02862-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2023] [Indexed: 07/20/2023]
Abstract
Coronary microvascular endothelial cells (CMECs) react to changes in coronary blood flow and myocardial metabolites and regulate coronary blood flow by balancing vasoconstrictors-such as endothelin-1-and the vessel dilators prostaglandin, nitric oxide, and endothelium-dependent hyperpolarizing factor. Coronary microvascular endothelial cell dysfunction is caused by several cardiovascular risk factors and chronic rheumatic diseases that impact CMEC blood flow regulation, resulting in coronary microcirculation dysfunction (CMD). The mechanisms of CMEC dysfunction are not fully understood. However, the following could be important mechanisms: the overexpression and activation of nicotinamide adenine dinucleotide phosphate oxidase (Nox), and mineralocorticoid receptors; the involvement of reactive oxygen species (ROS) caused by a decreased expression of sirtuins (SIRT3/SIRT1); forkhead box O3; and a decreased SKCA/IKCA expression in the endothelium-dependent hyperpolarizing factor electrical signal pathway. In addition, p66Shc is an adapter protein that promotes oxidative stress; although there are no studies on its involvement with cardiac microvessels, it is possible it plays an important role in CMD.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Department of Cardiology, The First Hospital of Jilin University, No. 71 of Xinmin Street, Changchun, 13000, China
| | - Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, College of Pharmacy, Jilin University, Changchun, 130000, China
| | - Jiahuan He
- Department of Cardiology, The First Hospital of Jilin University, No. 71 of Xinmin Street, Changchun, 13000, China
| | - Shipeng Wang
- Department of Cardiology, The First Hospital of Jilin University, No. 71 of Xinmin Street, Changchun, 13000, China
| | - Jingyue Wang
- Department of Cardiology, The First Hospital of Jilin University, No. 71 of Xinmin Street, Changchun, 13000, China
| | - Junqian Liu
- Department of Cardiology, The First Hospital of Jilin University, No. 71 of Xinmin Street, Changchun, 13000, China
| | - Yushi Wang
- Department of Cardiology, The First Hospital of Jilin University, No. 71 of Xinmin Street, Changchun, 13000, China.
| |
Collapse
|
14
|
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
|
15
|
Peh ZH, Dihoum A, Hutton D, Arthur JSC, Rena G, Khan F, Lang CC, Mordi IR. Inflammation as a therapeutic target in heart failure with preserved ejection fraction. Front Cardiovasc Med 2023; 10:1125687. [PMID: 37456816 PMCID: PMC10339321 DOI: 10.3389/fcvm.2023.1125687] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for around half of all cases of heart failure and may become the dominant type of heart failure in the near future. Unlike HF with reduced ejection fraction there are few evidence-based treatment strategies available. There is a significant unmet need for new strategies to improve clinical outcomes in HFpEF patients. Inflammation is widely thought to play a key role in HFpEF pathophysiology and may represent a viable treatment target. In this review focusing predominantly on clinical studies, we will summarise the role of inflammation in HFpEF and discuss potential therapeutic strategies targeting inflammation.
Collapse
Affiliation(s)
- Zhen Hui Peh
- School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - Adel Dihoum
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Dana Hutton
- School of Medicine, University of Dundee, Ninewells Hospital, Dundee, United Kingdom
| | - J. Simon C. Arthur
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Graham Rena
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Faisel Khan
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Chim C. Lang
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Ify R. Mordi
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
16
|
Shah SA, Reagan CE, Bresticker JE, Wolpe AG, Good ME, Macal EH, Billcheck HO, Bradley LA, French BA, Isakson BE, Wolf MJ, Epstein FH. Obesity-Induced Coronary Microvascular Disease Is Prevented by iNOS Deletion and Reversed by iNOS Inhibition. JACC Basic Transl Sci 2023; 8:501-514. [PMID: 37325396 PMCID: PMC10264569 DOI: 10.1016/j.jacbts.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 02/04/2023]
Abstract
Coronary microvascular disease (CMD) caused by obesity and diabetes is major contributor to heart failure with preserved ejection fraction; however, the mechanisms underlying CMD are not well understood. Using cardiac magnetic resonance applied to mice fed a high-fat, high-sucrose diet as a model of CMD, we elucidated the role of inducible nitric oxide synthase (iNOS) and 1400W, an iNOS antagonist, in CMD. Global iNOS deletion prevented CMD along with the associated oxidative stress and diastolic and subclinical systolic dysfunction. The 1400W treatment reversed established CMD and oxidative stress and preserved systolic/diastolic function in mice fed a high-fat, high-sucrose diet. Thus, iNOS may represent a therapeutic target for CMD.
Collapse
Affiliation(s)
- Soham A. Shah
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Claire E. Reagan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Julia E. Bresticker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Abigail G. Wolpe
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Miranda E. Good
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Edgar H. Macal
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| | - Helen O. Billcheck
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Leigh A. Bradley
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Brent A. French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Brant E. Isakson
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia, USA
| | - Matthew J. Wolf
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
- Department of Cardiovascular Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Frederick H. Epstein
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- The Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
17
|
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
|
18
|
Regidor PA, Mueller A, Mayr M. Pharmacological and metabolic effects of drospirenone as a progestin-only pill compared to combined formulations with estrogen. WOMEN'S HEALTH (LONDON, ENGLAND) 2023; 19:17455057221147388. [PMID: 36744531 PMCID: PMC9905034 DOI: 10.1177/17455057221147388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The spironolactone derivative drospirenone is combined with ethinylestradiol or estetrol in combined oral contraceptives. Formulations with 17-β-estradiol are used to treat climacteric symptoms. A drospirenone-only formulation has been introduced for contraception. Here, the pharmacological properties of drospirenone, the impact of the different formulations on metabolic and laboratory parameters, and the resulting clinical implications are reviewed. Ethinylestradiol, an inhibitor of CYP metabolic enzymes, changes the pharmacokinetics of drospirenone, leading to a higher drospirenone exposure with ethinylestradiol/drospirenone compared to the drospirenone-only preparation. In addition, several metabolic alterations have been described. The impact of estetrol is less pronounced, and for 17-β-estradiol/drospirenone and drospirenone-only, decreased triglyceride and cholesterol levels were observed. Ethinylestradiol induces various pro-coagulatory factors, leading to hypercoagulability. The effect is significantly reduced with estetrol, and no influence was observed with the drospirenone-only preparation. The anti-mineralocorticoid activity of drospirenone seems to positively counteract the renin-angiotensin-aldosterone-system-activating action of ethinylestradiol. There is no influence on blood pressure with ethinylestradiol/drospirenone and estetrol/drospirenone formulations, while in clinical trials, a reduction has been observed with 17-β-estradiol/drospirenone and drospirenone-only. Anti-aldosterone activity via non-renal mineralocorticoid receptors is associated with cardiovascular health, while interactions with parathyroid hormone signaling impact bone structure and vascular calcification. Though the clinical relevance is unclear for drospirenone, data in this context are reviewed. To sum up, the advantages of drospirenone in hormonal contraception and treatment of menopausal symptoms have been demonstrated for all the formulations described here. Combination with estrogen confers benefits and risks, which must be considered.
Collapse
Affiliation(s)
- Pedro-Antonio Regidor
- Pedro-Antonio Regidor, Exeltis Germany GmbH, Adalperostraße 84, 85737 Ismaning, Germany.
| | | | | |
Collapse
|
19
|
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
|
20
|
D’Elia JA, Bayliss GP, Weinrauch LA. The Diabetic Cardiorenal Nexus. Int J Mol Sci 2022; 23:ijms23137351. [PMID: 35806355 PMCID: PMC9266839 DOI: 10.3390/ijms23137351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/10/2022] Open
Abstract
The end-stage of the clinical combination of heart failure and kidney disease has become known as cardiorenal syndrome. Adverse consequences related to diabetes, hyperlipidemia, obesity, hypertension and renal impairment on cardiovascular function, morbidity and mortality are well known. Guidelines for the treatment of these risk factors have led to the improved prognosis of patients with coronary artery disease and reduced ejection fraction. Heart failure hospital admissions and readmission often occur, however, in the presence of metabolic, renal dysfunction and relatively preserved systolic function. In this domain, few advances have been described. Diabetes, kidney and cardiac dysfunction act synergistically to magnify healthcare costs. Current therapy relies on improving hemodynamic factors destructive to both the heart and kidney. We consider that additional hemodynamic solutions may be limited without the use of animal models focusing on the cardiomyocyte, nephron and extracellular matrices. We review herein potential common pathophysiologic targets for treatment to prevent and ameliorate this syndrome.
Collapse
Affiliation(s)
- John A. D’Elia
- Kidney and Hypertension Section, E P Joslin Research Laboratory, Joslin Diabetes Center, Boston, MA 02215, USA
| | - George P. Bayliss
- Division of Organ Transplantation, Rhode Island Hospital, Providence, RI 02903, USA;
| | - Larry A. Weinrauch
- Kidney and Hypertension Section, E P Joslin Research Laboratory, Joslin Diabetes Center, Boston, MA 02215, USA
- Correspondence: ; Tel.: +617-923-0800; Fax: +617-926-5665
| |
Collapse
|
21
|
Patil S, Rojulpote C, Amanullah A. Primary Aldosteronism and Ischemic Heart Disease. Front Cardiovasc Med 2022; 9:882330. [PMID: 35677685 PMCID: PMC9168042 DOI: 10.3389/fcvm.2022.882330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular disease, in particular ischemic heart disease is a major cause of morbidity and mortality worldwide. Primary aldosteronism is the leading cause of secondary hypertension, yet commonly under diagnosed, and represents a major preventable risk factor. In contrast to historical teaching, recent studies have shown that excess aldosterone production is associated with increased burden of ischemic heart disease disproportionate to the effects caused by hypertension alone. Aldosterone through its genomic and non-genomic actions exerts various detrimental cardiovascular changes contributing to this elevated risk. Recognition of primary hyperaldosteronism and understanding the distinctive pathophysiology of ischemic heart disease in primary aldosteronism is crucial to develop strategies to improve outcomes.
Collapse
Affiliation(s)
- Shivaraj Patil
- Department of Cardiology, Einstein Medical Center, Philadelphia, PA, United States
| | - Chaitanya Rojulpote
- Department of Medicine, The Wright Center for Graduate Medical Education, Scranton, PA, United States
| | - Aman Amanullah
- Department of Cardiology, Einstein Medical Center, Philadelphia, PA, United States
| |
Collapse
|
22
|
Abstract
Purpose of Review The regulation of blood pressure is conventionally conceptualised into the product of “circulating blood volume” and “vasoconstriction components”. Over the last few years, however, demonstration of tissue sodium storage challenged this dichotomous view. Recent Findings We review the available evidence pertaining to this phenomenon and the early association made with blood pressure; we discuss open questions regarding its originally proposed hypertonic nature, recently challenged by the suggestion of a systemic, isotonic, water paralleled accumulation that mirrors absolute or relative extracellular volume expansion; we present the established and speculate on the putative implications of this extravascular sodium excess, in either volume-associated or -independent form, on blood pressure regulation; finally, we highlight the prevalence of high tissue sodium in cardiovascular, metabolic and inflammatory conditions other than hypertension. Summary We conclude on approaches to reduce sodium excess and on the potential of emerging imaging technologies in hypertension and other conditions.
Collapse
|
23
|
Henry TD, Bairey Merz CN, Wei J, Corban MT, Quesada O, Joung S, Kotynski CL, Wang J, Lewis M, Schumacher AM, Bartel RL, Takagi H, Shah V, Lee A, Sietsema WK, Losordo DW, Lerman A. Autologous CD34+ Stem Cell Therapy Increases Coronary Flow Reserve and Reduces Angina in Patients With Coronary Microvascular Dysfunction. Circ Cardiovasc Interv 2022; 15:e010802. [PMID: 35067072 PMCID: PMC8843403 DOI: 10.1161/circinterventions.121.010802] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 11/09/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Coronary microvascular dysfunction results in angina and adverse outcomes in patients with evidence of ischemia and nonobstructive coronary artery disease; however, no specific therapy exists. CD34+ cell therapy increases microvasculature in preclinical models and improves symptoms, exercise tolerance, and mortality in refractory angina patients with obstructive coronary artery disease. The objective of this research was to evaluate the safety, tolerability, and efficacy of intracoronary CD34+ cell therapy in patients with coronary microvascular dysfunction. METHODS We conducted a 2-center, 20-participant trial of autologous CD34+ cell therapy (protocol CLBS16-P01; NCT03508609) in patients with ischemia and nonobstructive coronary artery disease with persistent angina and coronary flow reserve ≤2.5. Efficacy measures included coronary flow reserve, angina frequency, Canadian Cardiovascular Society angina class, Seattle Angina Questionnaire, SF-36, and modified Bruce exercise treadmill test obtained at baseline and 6 months after treatment. Autologous CD34+ cells (CLBS16) were mobilized by administration of granulocyte-colony stimulating factor 5µg/kg/day for 5 days and collected by leukapheresis. Participants received a single intracoronary left anterior descending infusion of isolated CD34+ cells in medium that enhances cell function. RESULTS Coronary flow reserve improved from 2.08±0.32 at baseline to 2.68±0.79 at 6 months after treatment (P<0.005). Angina frequency decreased (P<0.004), Canadian Cardiovascular Society class improved (P<0.001), and quality of life improved as assessed by the Seattle Angina Questionnaire (P≤0.03, all scales) and SF-36 (P≤0.04, all scales). There were no cell-related serious adverse events. CONCLUSIONS In this pilot clinical trial of microvascular angina, patients with ischemia and nonobstructive coronary artery disease receiving intracoronary infusion of CD34+ cell therapy had higher coronary flow reserve, less severe angina, and better quality of life at 6 months. The current study supports a potential therapeutic role for CD34+ cells in patients with microvascular angina. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03508609.
Collapse
Affiliation(s)
- Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital, Cincinnati, OH (O.Q., T.D.H.)
| | - C. Noel Bairey Merz
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (C.N.B.M., J.W., S.J.)
| | - Janet Wei
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (C.N.B.M., J.W., S.J.)
| | | | - Odayme Quesada
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital, Cincinnati, OH (O.Q., T.D.H.)
| | - Sandy Joung
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (C.N.B.M., J.W., S.J.)
| | - Christine L. Kotynski
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Jian Wang
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Michelle Lewis
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Ann M. Schumacher
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Ronnda L. Bartel
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Hiroshi Takagi
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Vishal Shah
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Anna Lee
- Mayo Clinic, Rochester, MN (M.T.C., A.L.)
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - William K. Sietsema
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Douglas W. Losordo
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| | - Amir Lerman
- Caladrius Biosciences, Basking Ridge, NJ (C.L.K., J.W., M.L., A.M.S., R.L.B., H.T., V.S., A.L., W.K.S., D.W.L.)
| |
Collapse
|
24
|
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
|
25
|
Srinivasa S, Thomas TS, Feldpausch MN, Adler GK, Grinspoon SK. Coronary Vasculature and Myocardial Structure in HIV: Physiologic Insights From the Renin-Angiotensin-Aldosterone System. J Clin Endocrinol Metab 2021; 106:3398-3412. [PMID: 33624807 PMCID: PMC8864747 DOI: 10.1210/clinem/dgab112] [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: 10/30/2020] [Indexed: 11/19/2022]
Abstract
The landscape of HIV medicine dramatically changed with the advent of contemporary antiretroviral therapies, which has allowed persons with HIV (PWH) to achieve good virologic control, essentially eliminating HIV-related complications and increasing life expectancy. As PWH are living longer, noncommunicable diseases, such as cardiovascular disease (CVD), have become a leading cause of morbidity and mortality in PWH with rates that are 50% to 100% higher than in well-matched persons without HIV. In this review, we focus on disease of the coronary microvasculature and myocardium in HIV. We highlight a key hormonal system important to cardiovascular endocrinology, the renin-angiotensin-aldosterone system (RAAS), as a potential mediator of inflammatory driven-vascular and myocardial injury and consider RAAS blockade as a physiologically targeted strategy to reduce CVD in HIV.
Collapse
Affiliation(s)
- Suman Srinivasa
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Teressa S Thomas
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Meghan N Feldpausch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gail K Adler
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Correspondence: Steven K. Grinspoon, MD, Metabolism Unit, Massachusetts General Hospital, 55 Fruit Street, 5LON207, Boston, MA 02114, USA. E-mail:
| |
Collapse
|
26
|
Ya'qoub L, Elgendy IY, Pepine CJ. Syndrome of Nonobstructive Coronary Artery Diseases: A Comprehensive Overview of Open Artery Ischemia. Am J Med 2021; 134:1321-1329. [PMID: 34343507 PMCID: PMC8754003 DOI: 10.1016/j.amjmed.2021.06.038] [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: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023]
Abstract
Syndromes of cardiac ischemia with nonobstructive coronary arteries have been increasingly recognized as a clinical entity with heterogeneous clinical presentations, commonly encountered in women. Knowledge of pathophysiology and clinical risk factors is key to ensuring appropriate diagnostic evaluation and management for these often-neglected patients. In this review, we discuss the epidemiology, risk factors, and clinical presentations of these syndromes. We provide algorithms for diagnosis and management of these entities based on current scientific knowledge and highlight some of the key knowledge gaps and ongoing trials in this emerging field.
Collapse
Affiliation(s)
- Lina Ya'qoub
- Department of Cardiology, Ochsner-Louisiana State University, Shreveport, La
| | - Islam Y Elgendy
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Carl J Pepine
- Division of Cardiovascular Medicine, University of Florida, Gainesville.
| |
Collapse
|
27
|
Lewis GA, Dodd S, Clayton D, Bedson E, Eccleson H, Schelbert EB, Naish JH, Jimenez BD, Williams SG, Cunnington C, Ahmed FZ, Cooper A, Rajavarma Viswesvaraiah, Russell S, McDonagh T, Williamson PR, Miller CA. Pirfenidone in heart failure with preserved ejection fraction: a randomized phase 2 trial. Nat Med 2021; 27:1477-1482. [PMID: 34385704 DOI: 10.1038/s41591-021-01452-0] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 06/25/2021] [Indexed: 11/09/2022]
Abstract
In heart failure with preserved ejection fraction (HFpEF), the occurrence of myocardial fibrosis is associated with adverse outcome. Whether pirfenidone, an oral antifibrotic agent without hemodynamic effect, is efficacious and safe for the treatment of HFpEF is unknown. In this double-blind, phase 2 trial ( NCT02932566 ), we enrolled patients with heart failure, an ejection fraction of 45% or higher and elevated levels of natriuretic peptides. Eligible patients underwent cardiovascular magnetic resonance and those with evidence of myocardial fibrosis, defined as a myocardial extracellular volume of 27% or greater, were randomly assigned to receive pirfenidone or placebo for 52 weeks. Forty-seven patients were randomized to each of the pirfenidone and placebo groups. The primary outcome was change in myocardial extracellular volume, from baseline to 52 weeks. In comparison to placebo, pirfenidone reduced myocardial extracellular volume (between-group difference, -1.21%; 95% confidence interval, -2.12 to -0.31; P = 0.009), meeting the predefined primary outcome. Twelve patients (26%) in the pirfenidone group and 14 patients (30%) in the placebo group experienced one or more serious adverse events. The most common adverse events in the pirfenidone group were nausea, insomnia and rash. In conclusion, among patients with HFpEF and myocardial fibrosis, administration of pirfenidone for 52 weeks reduced myocardial fibrosis. The favorable effects of pirfenidone in patients with HFpEF will need to be confirmed in future trials.
Collapse
Affiliation(s)
- Gavin A Lewis
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Susanna Dodd
- Department of Health Data Science, University of Liverpool, a Member of Liverpool Health Partners, Liverpool, UK
| | - Dannii Clayton
- Liverpool Clinical Trials Centre, University of Liverpool, Institute of Child Health, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Emma Bedson
- Liverpool Clinical Trials Centre, University of Liverpool, Institute of Child Health, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Helen Eccleson
- Liverpool Clinical Trials Centre, University of Liverpool, Institute of Child Health, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Erik B Schelbert
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Josephine H Naish
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | | | | | | | - Fozia Zahir Ahmed
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Anne Cooper
- Salford Royal NHS Foundation Trust, Salford, UK
| | | | | | | | - Paula R Williamson
- Department of Health Data Science, University of Liverpool, a Member of Liverpool Health Partners, Liverpool, UK
| | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
- Manchester University NHS Foundation Trust, Manchester, UK.
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Gadasheva Y, Nolze A, Grossmann C. Posttranslational Modifications of the Mineralocorticoid Receptor and Cardiovascular Aging. Front Mol Biosci 2021; 8:667990. [PMID: 34124152 PMCID: PMC8193679 DOI: 10.3389/fmolb.2021.667990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/07/2021] [Indexed: 01/05/2023] Open
Abstract
During aging, the cardiovascular system is especially prone to a decline in function and to life-expectancy limiting diseases. Cardiovascular aging is associated with increased arterial stiffness and vasoconstriction as well as left ventricular hypertrophy and reduced diastolic function. Pathological changes include endothelial dysfunction, atherosclerosis, fibrosis, hypertrophy, inflammation, and changes in micromilieu with increased production of reactive oxygen and nitrogen species. The renin-angiotensin-aldosterone-system is an important mediator of electrolyte and blood pressure homeostasis and a key contributor to pathological remodeling processes of the cardiovascular system. Its effects are partially conveyed by the mineralocorticoid receptor (MR), a ligand-dependent transcription factor, whose activity increases during aging and cardiovascular diseases without correlating changes of its ligand aldosterone. There is growing evidence that the MR can be enzymatically and non-enzymatically modified and that these modifications contribute to ligand-independent modulation of MR activity. Modifications reported so far include phosphorylation, acetylation, ubiquitination, sumoylation and changes induced by nitrosative and oxidative stress. This review focuses on the different posttranslational modifications of the MR, their impact on MR function and degradation and the possible implications for cardiovascular aging and diseases.
Collapse
Affiliation(s)
- Yekatarina Gadasheva
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Alexander Nolze
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
29
|
The Acute Effects of Different Spironolactone Doses on Oxidative Stress in Streptozotocin-Induced Diabetic Rats. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2021. [DOI: 10.2478/sjecr-2021-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality in patients with diabetes mellitus. Increased bioavailability of reactive oxygen species is defined as oxidative stress and is noticed in type 2 DM and reduced antioxidant enzymes expression/ activity. Aldosterone, an adrenal hormone, is secreted due to renin-angiotensin–aldosterone system activation, representing one of the fundamental physiological reactions in CVD. Spironolactone, a mineralocorticoid receptor antagonist, uses enhanced coronary microvascular function, suggesting a beneficial role of aldosterone in preventing diabetic cardiovascular complications in patients with type 2 DM. In this study, we evaluated the influence of spironolactone's acute administration on oxidative stress in rats with diabetes mellitus induced by streptozotocin. The present study was carried out on 40 adult male Wistar albino rats (8 weeks old). Rats were randomly divided into 4 groups (10 animals per group): healthy rats treated with 0.1 μM of spironolactone, diabetic rats treated with 0.1 μM of spironolactone, healthy rats treated with 3 μM of spironolactone, and diabetic rats treated with 3 μM of spironolactone. Spironolactone achieved different effects on oxidative stress parameters when given acutely in different doses in diabetic and healthy rats. In lower doses, spironolactone's acute administration reached lowered parameters of oxidative stress in healthy rats better than higher doses of spironolactone. In contrast, in the diabetic group, acute effects of higher doses of spironolactone lowered oxidative stress parameters better than lower spironolactone doses.
Collapse
|
30
|
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
|
31
|
Houben AJ, Stehouwer CD. Microvascular dysfunction: Determinants and treatment, with a focus on hyperglycemia. ENDOCRINE AND METABOLIC SCIENCE 2021. [DOI: 10.1016/j.endmts.2020.100073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
32
|
Kim SK, Biwer LA, Moss ME, Man JJ, Aronovitz MJ, Martin GL, Carrillo-Salinas FJ, Salvador AM, Alcaide P, Jaffe IZ. Mineralocorticoid Receptor in Smooth Muscle Contributes to Pressure Overload-Induced Heart Failure. Circ Heart Fail 2021; 14:e007279. [PMID: 33517669 PMCID: PMC7887087 DOI: 10.1161/circheartfailure.120.007279] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mineralocorticoid receptor (MR) antagonists decrease heart failure (HF) hospitalization and mortality, but the mechanisms are unknown. Preclinical studies reveal that the benefits on cardiac remodeling and dysfunction are not completely explained by inhibition of MR in cardiomyocytes, fibroblasts, or endothelial cells. The role of MR in smooth muscle cells (SMCs) in HF has never been explored. METHODS Male mice with inducible deletion of MR from SMCs (SMC-MR-knockout) and their MR-intact littermates were exposed to HF induced by 27-gauge transverse aortic constriction versus sham surgery. HF phenotypes and mechanisms were measured 4 weeks later using cardiac ultrasound, intracardiac pressure measurements, exercise testing, histology, cardiac gene expression, and leukocyte flow cytometry. RESULTS Deletion of MR from SMC attenuated transverse aortic constriction-induced HF with statistically significant improvements in ejection fraction, cardiac stiffness, chamber dimensions, intracardiac pressure, pulmonary edema, and exercise capacity. Mechanistically, SMC-MR-knockout protected from adverse cardiac remodeling as evidenced by decreased cardiomyocyte hypertrophy and fetal gene expression, interstitial and perivascular fibrosis, and inflammatory and fibrotic gene expression. Exposure to pressure overload resulted in a statistically significant decline in cardiac capillary density and coronary flow reserve in MR-intact mice. These vascular parameters were improved in SMC-MR-knockout mice compared with MR-intact littermates exposed to transverse aortic constriction. CONCLUSIONS These results provide a novel paradigm by which MR inhibition may be beneficial in HF by blocking MR in SMC, thereby improving cardiac blood supply in the setting of pressure overload-induced hypertrophy, which in turn mitigates the adverse cardiac remodeling that contributes to HF progression and symptoms.
Collapse
MESH Headings
- Animals
- Aorta/surgery
- Arterial Pressure
- Cardiomegaly/genetics
- Cardiomegaly/pathology
- Cardiomegaly/physiopathology
- Constriction, Pathologic
- Disease Models, Animal
- Echocardiography
- Gene Knockout Techniques
- Heart Failure/diagnostic imaging
- Heart Failure/genetics
- Heart Failure/pathology
- Heart Failure/physiopathology
- Mice
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/physiology
- Receptors, Mineralocorticoid/genetics
- Ventricular Remodeling/genetics
Collapse
Affiliation(s)
- Seung Kyum Kim
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
- Department of Sports Science, Seoul National University of Science and Technology, Seoul, Republic of Korea
| | - Lauren A. Biwer
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | - M. Elizabeth Moss
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Joshua J. Man
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA
| | - Mark J. Aronovitz
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | - Gregory L. Martin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| | | | - Ane M. Salvador
- Department of Immunology, Tufts University School of Medicine, Boston, MA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA
| | - Iris Z. Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA
| |
Collapse
|
33
|
Gulsin GS, Henson J, Brady EM, Sargeant JA, Wilmot EG, Athithan L, Htike ZZ, Marsh AM, Biglands JD, Kellman P, Khunti K, Webb D, Davies MJ, Yates T, McCann GP. Cardiovascular Determinants of Aerobic Exercise Capacity in Adults With Type 2 Diabetes. Diabetes Care 2020; 43:2248-2256. [PMID: 32680830 PMCID: PMC7440912 DOI: 10.2337/dc20-0706] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/09/2020] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To assess the relationship between subclinical cardiac dysfunction and aerobic exercise capacity (peak VO2) in adults with type 2 diabetes (T2D), a group at high risk of developing heart failure. RESEARCH DESIGN AND METHODS Cross-sectional study. We prospectively enrolled a multiethnic cohort of asymptomatic adults with T2D and no history, signs, or symptoms of cardiovascular disease. Age-, sex-, and ethnicity-matched control subjects were recruited for comparison. Participants underwent bioanthropometric profiling, cardiopulmonary exercise testing, and cardiovascular magnetic resonance with adenosine stress perfusion imaging. Multivariable linear regression analysis was undertaken to identify independent associations between measures of cardiovascular structure and function and peak VO2. RESULTS A total of 247 adults with T2D (aged 51.8 ± 11.9 years, 55% males, 37% black or south Asian ethnicity, HbA1c 7.4 ± 1.1% [57 ± 12 mmol/mol], and duration of diabetes 61 [32-120] months) and 78 control subjects were included. Subjects with T2D had increased concentric left ventricular remodeling, reduced myocardial perfusion reserve (MPR), and markedly lower aerobic exercise capacity (peak VO2 18.0 ± 6.6 vs. 27.8 ± 9.0 mL/kg/min; P < 0.001) compared with control subjects. In a multivariable linear regression model containing age, sex, ethnicity, smoking status, and systolic blood pressure, only MPR (β = 0.822; P = 0.006) and left ventricular diastolic filling pressure (E/e') (β = -0.388; P = 0.001) were independently associated with peak VO2 in subjects with T2D. CONCLUSIONS In a multiethnic cohort of asymptomatic people with T2D, MPR and diastolic function are key determinants of aerobic exercise capacity, independent of age, sex, ethnicity, smoking status, or blood pressure.
Collapse
Affiliation(s)
- Gaurav S Gulsin
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K.
| | - Joseph Henson
- Diabetes Research Centre, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Emer M Brady
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Jack A Sargeant
- Diabetes Research Centre, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Emma G Wilmot
- Diabetes Department, Royal Derby Hospital, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, U.K
| | - Lavanya Athithan
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Zin Z Htike
- Diabetes Department, Royal Derby Hospital, University Hospitals of Derby and Burton NHS Foundation Trust, Derby, U.K
| | - Anna-Marie Marsh
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | | | - Peter Kellman
- National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - David Webb
- Diabetes Research Centre, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Melanie J Davies
- Diabetes Research Centre, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Thomas Yates
- Diabetes Research Centre, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K
| | - Gerry P McCann
- Department of Cardiovascular Sciences, University of Leicester and the NIHR Leicester Biomedical Research Centre, Leicester, U.K.
| |
Collapse
|
34
|
Packer M, Lam CS, Lund LH, Maurer MS, Borlaug BA. Characterization of the inflammatory-metabolic phenotype of heart failure with a preserved ejection fraction: a hypothesis to explain influence of sex on the evolution and potential treatment of the disease. Eur J Heart Fail 2020; 22:1551-1567. [PMID: 32441863 PMCID: PMC7687188 DOI: 10.1002/ejhf.1902] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/05/2020] [Accepted: 05/17/2020] [Indexed: 12/11/2022] Open
Abstract
Accumulating evidence points to the existence of an inflammatory-metabolic phenotype of heart failure with a preserved ejection fraction (HFpEF), which is characterized by biomarkers of inflammation, an expanded epicardial adipose tissue mass, microvascular endothelial dysfunction, normal-to-mildly increased left ventricular volumes and systolic blood pressures, and possibly, altered activity of adipocyte-associated inflammatory mediators. A broad range of adipogenic metabolic and systemic inflammatory disorders - e.g. obesity, diabetes and metabolic syndrome as well as rheumatoid arthritis and psoriasis - can cause this phenotype, independent of the presence of large vessel coronary artery disease. Interestingly, when compared with men, women are both at greater risk of and may suffer greater cardiac consequences from these systemic inflammatory and metabolic disorders. Women show disproportionate increases in left ventricular filling pressures following increases in central blood volume and have greater arterial stiffness than men. Additionally, they are particularly predisposed to epicardial and intramyocardial fat expansion and imbalances in adipocyte-associated proinflammatory mediators. The hormonal interrelationships seen in inflammatory-metabolic phenotype may explain why mineralocorticoid receptor antagonists and neprilysin inhibitors may be more effective in women than in men with HFpEF. Recognition of the inflammatory-metabolic phenotype may improve an understanding of the pathogenesis of HFpEF and enhance the ability to design clinical trials of interventions in this heterogeneous syndrome.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular InstituteBaylor University Medical CenterDallasTXUSA
- Imperial College LondonLondonUK
| | - Carolyn S.P. Lam
- National Heart Centre Singapore and Duke‐National University of SingaporeSingapore
- University Medical Centre GroningenGroningenThe Netherlands
- The George Institute for Global HealthSydneyAustralia
| | - Lars H. Lund
- Department of Medicine, Karolinska Institutet and Heart and Vascular ThemeKarolinska University HospitalStockholmSweden
| | | | | |
Collapse
|
35
|
Zhou W, Brown JM, Bajaj NS, Chandra A, Divakaran S, Weber B, Bibbo CF, Hainer J, Taqueti VR, Dorbala S, Blankstein R, Adler D, O’Gara P, Di Carli MF. Hypertensive coronary microvascular dysfunction: a subclinical marker of end organ damage and heart failure. Eur Heart J 2020; 41:2366-2375. [PMID: 32221588 PMCID: PMC7327534 DOI: 10.1093/eurheartj/ehaa191] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/14/2020] [Accepted: 03/03/2020] [Indexed: 12/28/2022] Open
Abstract
AIMS Hypertension is a well-established heart failure (HF) risk factor, especially in the context of adverse left ventricular (LV) remodelling. We aimed to use myocardial flow reserve (MFR) and global longitudinal strain (GLS), markers of subclinical microvascular and myocardial dysfunction, to refine hypertensive HF risk assessment. METHODS AND RESULTS Consecutive patients undergoing symptom-prompted stress cardiac positron emission tomography (PET)-computed tomography and transthoracic echocardiogram within 90 days without reduced left ventricular ejection fraction (<40%) or flow-limiting coronary artery disease (summed stress score ≥ 3) were included. Global MFR was quantified by PET, and echocardiograms were retrospectively analysed for cardiac structure and function. Patients were followed over a median 8.75 (Q1-3 4.56-10.04) years for HF hospitalization and a composite of death, HF hospitalization, MI, or stroke. Of 194 patients, 155 had adaptive LV remodelling while 39 had maladaptive remodelling, which was associated with lower MFR and impaired GLS. Across the remodelling spectrum, diastolic parameters, GLS, and N-terminal pro-B-type natriuretic peptide were independently associated with MFR. Maladaptive LV remodelling was associated with increased adjusted incidence of HF hospitalization and death. Importantly, the combination of abnormal MFR and GLS was associated with a higher rate of HF hospitalization compared to normal MFR and GLS [adjusted hazard ratio (HR) 3.21, 95% confidence interval (CI) 1.09-9.45, P = 0.034), including in the adaptive remodelling subset (adjusted HR 3.93, 95% CI 1.14-13.56, P = 0.030). CONCLUSION We have demonstrated important associations between coronary microvascular dysfunction and myocardial mechanics that refine disease characterization and HF risk assessment of patients with hypertension based on subclinical target organ injury.
Collapse
Affiliation(s)
- Wunan Zhou
- Cardiology Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814, USA
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jenifer M Brown
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Navkaranbir S Bajaj
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alvin Chandra
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sanjay Divakaran
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Brittany Weber
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Courtney F Bibbo
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jon Hainer
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Viviany R Taqueti
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sharmila Dorbala
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ron Blankstein
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dale Adler
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick O’Gara
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marcelo F Di Carli
- Cardiovascular Imaging Program, Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Heart and Vascular Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
36
|
Sirtuin 3, Endothelial Metabolic Reprogramming, and Heart Failure With Preserved Ejection Fraction. J Cardiovasc Pharmacol 2020; 74:315-323. [PMID: 31425381 DOI: 10.1097/fjc.0000000000000719] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The incidences of heart failure with preserved ejection fraction (HFpEF) are increased in aged populations as well as diabetes and hypertension. Coronary microvascular dysfunction has contributed to the development of HFpEF. Endothelial cells (ECs) depend on glycolysis rather than oxidative phosphorylation for generating adenosine triphosphate to maintain vascular homeostasis. Glycolytic metabolism has a critical role in the process of angiogenesis, because ECs rely on the energy produced predominantly from glycolysis for migration and proliferation. Sirtuin 3 (SIRT3) is found predominantly in mitochondria and its expression declines progressively with aging, diabetes, obesity, and hypertension. Emerging evidence indicates that endothelial SIRT3 regulates a metabolic switch between glycolysis and mitochondrial respiration. SIRT3 deficiency in EC resulted in a significant decrease in glycolysis, whereas, it exhibited higher mitochondrial respiration and more prominent production of reactive oxygen species. SIRT3 deficiency also displayed striking increases in acetylation of p53, EC apoptosis, and senescence. Impairment of SIRT3-mediated EC metabolism may lead to a disruption of EC/pericyte/cardiomyocyte communications and coronary microvascular rarefaction, which promotes cardiomyocyte hypoxia, Titin-based cardiomyocyte stiffness, and myocardial fibrosis, thus leading to a diastolic dysfunction and HFpEF. This review summarizes current knowledge of SIRT3 in EC metabolic reprograming, EC/pericyte interactions, coronary microvascular dysfunction, and HFpEF.
Collapse
|
37
|
Hall JJ, Eurich DT, Nagy D, Tjosvold L, Gamble JM. Thiazide Diuretic-Induced Change in Fasting Plasma Glucose: a Meta-analysis of Randomized Clinical Trials. J Gen Intern Med 2020; 35:1849-1860. [PMID: 32157653 PMCID: PMC7280437 DOI: 10.1007/s11606-020-05731-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/10/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Prior meta-analyses measuring thiazide-induced glycemic change have demonstrated an increased risk of incident diabetes; however, this measure's definition has changed over time. AIM To determine the magnitude of change in fasting plasma glucose (FPG) for thiazide diuretics. DATA SOURCES A research librarian designed and conducted searches in Medline®, EMBASE, and EBM Reviews-Cochrane Central Register of Controlled Trials (inception through July 2018) and International Pharmaceutical Abstracts (inception to December 2014). STUDY SELECTION Randomized, controlled trials comparing a thiazide or thiazide-like diuretic to any comparator reporting FPG were identified. Trials enrolling < 50 participants, those with a follow-up period of < 4 weeks, and conference abstracts were excluded. DATA EXTRACTION Independent duplicate screening of citations and full-text articles, data extraction, and assessment of risk of bias was conducted. DATA SYNTHESIS Ninety-five studies were included (N = 76,608 participants), with thiazides compared with placebo, beta-blockers, calcium channel blockers, renin-angiotensin-aldosterone-system inhibitors, potassium-sparing diuretic, and others alone or in combination. Thiazide diuretics marginally increased FPG (weighted mean difference 0.20 mmol/L (95% CI 0.15-0.25); I2 = 84%) (1 mmol/L = 18 mg/dL). Results did not change substantially when considering dose or duration, comparing thiazides with placebo or an active comparator, or using thiazides as monotherapy or combination therapy, even when combined with a potassium-correcting agent. CONCLUSION Thiazide diuretics have a small and clinically unimportant impact on FPG.
Collapse
Affiliation(s)
- Jill J. Hall
- Faculty of Pharmacy and Pharmaceutical Sciences, 3-236 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB Canada
| | - Dean T. Eurich
- School of Public Health, University of Alberta, Edmonton, Canada
| | - Danielle Nagy
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Lisa Tjosvold
- John W. Scott Health Sciences Library, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
38
|
Yao PL, Peavey J, Malek G. Leveraging Nuclear Receptors as Targets for Pathological Ocular Vascular Diseases. Int J Mol Sci 2020; 21:ijms21082889. [PMID: 32326149 PMCID: PMC7215709 DOI: 10.3390/ijms21082889] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/18/2020] [Accepted: 04/19/2020] [Indexed: 02/07/2023] Open
Abstract
Vasculogenesis and angiogenesis are physiological mechanisms occurring throughout the body. Any disruption to the precise balance of blood vessel growth necessary to support healthy tissue, and the inhibition of abnormal vessel sprouting has the potential to negatively impact stages of development and/or healing. Therefore, the identification of key regulators of these vascular processes is critical to identifying therapeutic means by which to target vascular-associated compromises and complications. Nuclear receptors are a family of transcription factors that have been shown to be involved in modulating different aspects of vascular biology in many tissues systems. Most recently, the role of nuclear receptors in ocular biology and vasculopathies has garnered interest. Herein, we review studies that have used in vitro assays and in vivo models to investigate nuclear receptor-driven pathways in two ocular vascular diseases associated with blindness, wet or exudative age-related macular degeneration, and proliferative diabetic retinopathy. The potential therapeutic targeting of nuclear receptors for ocular diseases is also discussed.
Collapse
Affiliation(s)
- Pei-Li Yao
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27503, USA; (P.-L.Y.); (J.P.)
| | - Jeremy Peavey
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27503, USA; (P.-L.Y.); (J.P.)
| | - Goldis Malek
- Duke Eye Center, Department of Ophthalmology, Duke University School of Medicine, Durham, NC 27503, USA; (P.-L.Y.); (J.P.)
- Department of Pathology, Duke University School of Medicine, Durham, NC 27503, USA
- Correspondence: ; Tel.: +919-684-0820
| |
Collapse
|
39
|
Packer M. Characterization, Pathogenesis, and Clinical Implications of Inflammation-Related Atrial Myopathy as an Important Cause of Atrial Fibrillation. J Am Heart Assoc 2020; 9:e015343. [PMID: 32242478 PMCID: PMC7428644 DOI: 10.1161/jaha.119.015343] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Historically, atrial fibrillation has been observed in clinical settings of prolonged hemodynamic stress, eg, hypertension and valvular heart disease. However, recently, the most prominent precedents to atrial fibrillation are metabolic diseases that are associated with adipose tissue inflammation (ie, obesity and diabetes mellitus) and systemic inflammatory disorders (ie, rheumatoid arthritis and psoriasis). These patients typically have little evidence of left ventricular hypertrophy or dilatation; instead, imaging reveals abnormalities of the structure or function of the atria, particularly the left atrium, indicative of an atrial myopathy. The left atrium is enlarged, fibrotic and noncompliant, potentially because the predisposing disorder leads to an expansion of epicardial adipose tissue, which transmits proinflammatory mediators to the underlying left atrium. The development of an atrial myopathy not only leads to atrial fibrillation, but also contributes to pulmonary venous hypertension and systemic thromboembolism. These mechanisms explain why disorders of systemic or adipose tissue inflammation are accompanied an increased risk of atrial fibrillation, abnormalities of left atrium geometry and an enhanced risk of stroke. The risk of stroke exceeds that predicted by conventional cardiovascular risk factors or thromboembolism risk scores used to guide the use of anticoagulation, but it is strongly linked to clinical evidence and biomarkers of systemic inflammation.
Collapse
Affiliation(s)
- Milton Packer
- Baylor Heart and Vascular Institute Baylor University Medical Center Dallas TX.,Imperial College London United Kingdom
| |
Collapse
|
40
|
Feraco A, Marzolla V, Scuteri A, Armani A, Caprio M. Mineralocorticoid Receptors in Metabolic Syndrome: From Physiology to Disease. Trends Endocrinol Metab 2020; 31:205-217. [PMID: 31843490 DOI: 10.1016/j.tem.2019.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 01/28/2023]
Abstract
Over the past decade, several studies have shown that activity of extra-renal mineralocorticoid receptors (MR) regulates vascular tone, adipogenesis, adipose tissue function, and cardiomyocyte contraction. In mice, abnormal activation of MR in the vasculature and in adipose tissue favors the occurrence of several components of the metabolic syndrome (MetS), such as hypertension, obesity, and glucose intolerance. Accordingly, high levels of aldosterone are associated with obesity and MetS in humans, suggesting that altered activation of aldosterone-MR system in extra-renal tissues leads to profound metabolic dysfunctions. In this context, in addition to the classical indications for heart failure and hypertension, MR antagonists (MRAs) nowadays represent a promising approach to tackle cardiovascular and metabolic disorders occurring in the MetS.
Collapse
Affiliation(s)
- Alessandra Feraco
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Angelo Scuteri
- Department of Medical, Surgical, and Experimental Science, University of Sassari, Sassari, Italy
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Rome, Italy; Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Rome, Italy.
| |
Collapse
|
41
|
Sorop O, van de Wouw J, Chandler S, Ohanyan V, Tune JD, Chilian WM, Merkus D, Bender SB, Duncker DJ. Experimental animal models of coronary microvascular dysfunction. Cardiovasc Res 2020; 116:756-770. [PMID: 31926020 PMCID: PMC7061277 DOI: 10.1093/cvr/cvaa002] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/25/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022] Open
Abstract
Coronary microvascular dysfunction (CMD) is commonly present in patients with metabolic derangements and is increasingly recognized as an important contributor to myocardial ischaemia, both in the presence and absence of epicardial coronary atherosclerosis. The latter condition is termed 'ischaemia and no obstructive coronary artery disease' (INOCA). Notwithstanding the high prevalence of INOCA, effective treatment remains elusive. Although to date there is no animal model for INOCA, animal models of CMD, one of the hallmarks of INOCA, offer excellent test models for enhancing our understanding of the pathophysiology of CMD and for investigating novel therapies. This article presents an overview of currently available experimental models of CMD-with an emphasis on metabolic derangements as risk factors-in dogs, swine, rabbits, rats, and mice. In all available animal models, metabolic derangements are most often induced by a high-fat diet (HFD) and/or diabetes mellitus via injection of alloxan or streptozotocin, but there is also a wide variety of spontaneous as well as transgenic animal models which develop metabolic derangements. Depending on the number, severity, and duration of exposure to risk factors-all these animal models show perturbations in coronary microvascular (endothelial) function and structure, similar to what has been observed in patients with INOCA and comorbid conditions. The use of these animal models will be instrumental in identifying novel therapeutic targets and for the subsequent development and testing of novel therapeutic interventions to combat ischaemic heart disease, the number one cause of death worldwide.
Collapse
Affiliation(s)
- Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Jens van de Wouw
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Selena Chandler
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Johnathan D Tune
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, USA
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
- Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Marchioninistr. 27, 81377 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), 81377 Munich, Germany
| | - Shawn B Bender
- Department of Biomedical Sciences, 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
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
42
|
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
|
43
|
Considerations for Clinical Trials Targeting the Myocardial Interstitium. JACC Cardiovasc Imaging 2019; 12:2319-2331. [DOI: 10.1016/j.jcmg.2019.03.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 01/23/2023]
|
44
|
Athithan L, Gulsin GS, McCann GP, Levelt E. Diabetic cardiomyopathy: Pathophysiology, theories and evidence to date. World J Diabetes 2019; 10:490-510. [PMID: 31641426 PMCID: PMC6801309 DOI: 10.4239/wjd.v10.i10.490] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 02/05/2023] Open
Abstract
The prevalence of type 2 diabetes (T2D) has increased worldwide and doubled over the last two decades. It features among the top 10 causes of mortality and morbidity in the world. Cardiovascular disease is the leading cause of complications in diabetes and within this, heart failure has been shown to be the leading cause of emergency admissions in the United Kingdom. There are many hypotheses and well-evidenced mechanisms by which diabetic cardiomyopathy as an entity develops. This review aims to give an overview of these mechanisms, with particular emphasis on metabolic inflexibility. T2D is associated with inefficient substrate utilisation, an inability to increase glucose metabolism and dependence on fatty acid oxidation within the diabetic heart resulting in mitochondrial uncoupling, glucotoxicity, lipotoxicity and initially subclinical cardiac dysfunction and finally in overt heart failure. The review also gives a concise update on developments within clinical imaging, specifically cardiac magnetic resonance studies to characterise and phenotype early cardiac dysfunction in T2D. A better understanding of the pathophysiology involved provides a platform for targeted therapy in diabetes to prevent the development of early heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Lavanya Athithan
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Gerald P McCann
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom
| | - Eylem Levelt
- Multidisciplinary Cardiovascular Research Centre and Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LF9 7TF, United Kingdom
| |
Collapse
|
45
|
Barrera-Chimal J, Jaisser F. Vascular mineralocorticoid receptor activation and disease. Exp Eye Res 2019; 188:107796. [PMID: 31521629 DOI: 10.1016/j.exer.2019.107796] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022]
Abstract
Mineralocorticoid receptor activation in endothelial and smooth muscle cells can promote vascular disease by increasing oxidative stress, promoting inflammation, accelerating vascular stiffness, remodeling, and calcification, altering vessel responsiveness to various vasoactive factors, thus altering vascular tone and blood pressure, and by altering angiogenesis. Here, we review the recent evidence highlighting the impact of vascular mineralocorticoid receptor activation in pathological situations, including kidney injury, vascular injury associated with metabolic diseases, atherosclerosis, cerebral vascular injury during hypertension, vascular stiffening and aging, pulmonary hypertension, vascular calcification, cardiac remodeling, wound healing, inflammation, thrombosis, and disorders related to angiogenic defects in the eye. The possible mechanisms implicating mineralocorticoid receptor activation in various vascular disorders are discussed. Altogether, recent evidence points towards pharmacological mineralocorticoid receptor inhibition as a strategy to treat diseases in which overactivation of the mineralocorticoid receptor in endothelial and/or smooth muscle cells may play a pivotal role.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Medicina Traslacional, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México and Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Frederic Jaisser
- INSERM U1116, Clinical Investigation Centre, Lorraine University, Vandoeuvre-lès-Nancy, France; INI-CRCT (Cardiovascular and Renal Clinical Trialists) F-CRIN Network, Nancy, France; Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006, Paris, France.
| |
Collapse
|
46
|
van de Wouw J, Broekhuizen M, Sorop O, Joles JA, Verhaar MC, Duncker DJ, Danser AHJ, Merkus D. Chronic Kidney Disease as a Risk Factor for Heart Failure With Preserved Ejection Fraction: A Focus on Microcirculatory Factors and Therapeutic Targets. Front Physiol 2019; 10:1108. [PMID: 31551803 PMCID: PMC6737277 DOI: 10.3389/fphys.2019.01108] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF) and chronic kidney disease (CKD) co-exist, and it is estimated that about 50% of HF patients suffer from CKD. Although studies have been performed on the association between CKD and HF with reduced ejection fraction (HFrEF), less is known about the link between CKD and heart failure with preserved ejection fraction (HFpEF). Approximately, 50% of all patients with HF suffer from HFpEF, and this percentage is projected to rise in the coming years. Therapies for HFrEF are long established and considered quite successful. In contrast, clinical trials for treatment of HFpEF have all shown negative or disputable results. This is likely due to the multifactorial character and the lack of pathophysiological knowledge of HFpEF. The typical co-existence of HFpEF and CKD is partially due to common underlying comorbidities, such as hypertension, dyslipidemia and diabetes. Macrovascular changes accompanying CKD, such as hypertension and arterial stiffening, have been described to contribute to HFpEF development. Furthermore, several renal factors have a direct impact on the heart and/or coronary microvasculature and may underlie the association between CKD and HFpEF. These factors include: (1) activation of the renin-angiotensin-aldosterone system, (2) anemia, (3) hypercalcemia, hyperphosphatemia and increased levels of FGF-23, and (4) uremic toxins. This review critically discusses the above factors, focusing on their potential contribution to coronary dysfunction, left ventricular stiffening, and delayed left ventricular relaxation. We further summarize the directions of novel treatment options for HFpEF based on the contribution of these renal drivers.
Collapse
Affiliation(s)
- Jens van de Wouw
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Michelle Broekhuizen
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands.,Division of Neonatology, Department of Pediatrics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Oana Sorop
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - A H Jan Danser
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
47
|
Suhrs HE, Michelsen MM, Prescott E. Treatment strategies in coronary microvascular dysfunction: A systematic review of interventional studies. Microcirculation 2019; 26:e12430. [PMID: 29130567 DOI: 10.1111/micc.12430] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/06/2017] [Indexed: 12/12/2022]
Abstract
CMD has been associated with a wide spectrum of diseases and conditions, and it has proven to be a strong prognostic marker of morbidity and mortality. Despite increased attention, guideline-based treatment recommendations are lacking. We performed a systematic review of pharmacological and nonpharmacological interventions to improve coronary perfusion, assessed by IC Doppler, TTDE, PET, CMRI, transthoracic contrast perfusion echocardiography, and dilution techniques. No restrictions were made regarding the study design (randomized, placebo-controlled/randomized with active comparators/nonrandomized with or without a control group), the cardiac condition studied, or the coronary microvascular function at baseline. An electronic database search yielded 4485 records of which 80 studies met our inclusion criteria. Included studies were sorted according to intervention and study design. Studies were small and heterogeneous in methodology, and only few were placebo-controlled. Although some treatments looked promising, we found that no specific treatment was sufficiently well documented to be recommended in any patient groups. There is a need for larger well-designed clinical trials, and we suggest that future studies stratify study populations according to pathogenic mechanisms, thereby investigating whether an individualized treatment approach would be more successful.
Collapse
Affiliation(s)
- Hannah E Suhrs
- Department of Cardiology, Bispebjerg University Hospital, Copenhagen NV, Denmark
| | - Marie M Michelsen
- Department of Cardiology, Bispebjerg University Hospital, Copenhagen NV, Denmark
| | - Eva Prescott
- Department of Cardiology, Bispebjerg University Hospital, Copenhagen NV, Denmark
| |
Collapse
|
48
|
Abstract
Heart failure and related morbidity and mortality are increasing at an alarming rate, in large part, because of increases in aging, obesity, and diabetes mellitus. The clinical outcomes associated with heart failure are considerably worse for patients with diabetes mellitus than for those without diabetes mellitus. In people with diabetes mellitus, the presence of myocardial dysfunction in the absence of overt clinical coronary artery disease, valvular disease, and other conventional cardiovascular risk factors, such as hypertension and dyslipidemia, has led to the descriptive terminology, diabetic cardiomyopathy. The prevalence of diabetic cardiomyopathy is increasing in parallel with the increase in diabetes mellitus. Diabetic cardiomyopathy is initially characterized by myocardial fibrosis, dysfunctional remodeling, and associated diastolic dysfunction, later by systolic dysfunction, and eventually by clinical heart failure. Impaired cardiac insulin metabolic signaling, mitochondrial dysfunction, increases in oxidative stress, reduced nitric oxide bioavailability, elevations in advanced glycation end products and collagen-based cardiomyocyte and extracellular matrix stiffness, impaired mitochondrial and cardiomyocyte calcium handling, inflammation, renin-angiotensin-aldosterone system activation, cardiac autonomic neuropathy, endoplasmic reticulum stress, microvascular dysfunction, and a myriad of cardiac metabolic abnormalities have all been implicated in the development and progression of diabetic cardiomyopathy. Molecular mechanisms linked to the underlying pathophysiological changes include abnormalities in AMP-activated protein kinase, peroxisome proliferator-activated receptors, O-linked N-acetylglucosamine, protein kinase C, microRNA, and exosome pathways. The aim of this review is to provide a contemporary view of these instigators of diabetic cardiomyopathy, as well as mechanistically based strategies for the prevention and treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Guanghong Jia
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.)
| | - Michael A Hill
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.)
| | - James R Sowers
- From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.), University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.).
| |
Collapse
|
49
|
Lewis GA, Schelbert EB, Naish JH, Bedson E, Dodd S, Eccleson H, Clayton D, Jimenez BD, McDonagh T, Williams SG, Cooper A, Cunnington C, Ahmed FZ, Viswesvaraiah R, Russell S, Neubauer S, Williamson PR, Miller CA. Pirfenidone in Heart Failure with Preserved Ejection Fraction-Rationale and Design of the PIROUETTE Trial. Cardiovasc Drugs Ther 2019; 33:461-470. [PMID: 31069575 PMCID: PMC6689029 DOI: 10.1007/s10557-019-06876-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The PIROUETTE (PIRfenidOne in patients with heart failUre and preserved lEfT venTricular Ejection fraction) trial is designed to evaluate the efficacy and safety of the anti-fibrotic pirfenidone in patients with chronic heart failure and preserved ejection fraction (HFpEF) and myocardial fibrosis. HFpEF is a diverse syndrome associated with substantial morbidity and mortality. Myocardial fibrosis is a key pathophysiological mechanism of HFpEF and myocardial fibrotic burden is strongly and independently associated with adverse outcome. Pirfenidone is an oral anti-fibrotic agent, without haemodynamic effect, that leads to regression of myocardial fibrosis in preclinical models. It has proven clinical effectiveness in pulmonary fibrosis. METHODS The PIROUETTE trial is a randomised, double-blind, placebo-controlled phase II trial evaluating the efficacy and safety of 52 weeks of treatment with pirfenidone in patients with chronic HFpEF (symptoms and signs of heart failure, left ventricular ejection fraction ≥ 45%, elevated natriuretic peptides [BNP ≥ 100 pg/ml or NT-proBNP ≥ 300 pg/ml; or BNP ≥ 300 pg/ml or NT-proBNP ≥ 900 pg/ml if in atrial fibrillation]) and myocardial fibrosis (extracellular matrix (ECM) volume ≥ 27% measured using cardiovascular magnetic resonance). The primary outcome measure is change in myocardial ECM volume. A sub-study will investigate the relationship between myocardial fibrosis and myocardial energetics, and the impact of pirfenidone, using 31phosphorus magnetic resonance spectroscopy. DISCUSSION PIROUETTE will determine whether pirfenidone is superior to placebo in relation to regression of myocardial fibrosis and improvement in myocardial energetics in patients with HFpEF and myocardial fibrosis (NCT02932566). CLINICAL TRIAL REGISTRATION clinicaltrials.gov (NCT02932566) https://clinicaltrials.gov/ct2/show/NCT02932566.
Collapse
Affiliation(s)
- Gavin A Lewis
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Erik B Schelbert
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Josephine H Naish
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Emma Bedson
- Clinical Trials Research Centre, 2nd Floor - Institute in the Park, Alder Hey Children's NHS Foundation Trust, University of Liverpool, Member of Liverpool Health Partners, Liverpool, L12 2AP, UK
| | - Susanna Dodd
- Department of Biostatistics, University of Liverpool, Member of Liverpool Health Partners, Block F, Waterhouse Bld, 1-5 Brownlow Street, Liverpool, L69 3GL, UK
| | - Helen Eccleson
- Clinical Trials Research Centre, 2nd Floor - Institute in the Park, Alder Hey Children's NHS Foundation Trust, University of Liverpool, Member of Liverpool Health Partners, Liverpool, L12 2AP, UK
| | - Dannii Clayton
- Clinical Trials Research Centre, 2nd Floor - Institute in the Park, Alder Hey Children's NHS Foundation Trust, University of Liverpool, Member of Liverpool Health Partners, Liverpool, L12 2AP, UK
| | - Beatriz Duran Jimenez
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | | | - Simon G Williams
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Anne Cooper
- Salford Royal NHS Foundation Trust, Stott Lane, Salford, M6 8HD, UK
| | - Colin Cunnington
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Fozia Zahir Ahmed
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK
| | - Rajavarma Viswesvaraiah
- Stockport NHS Foundation Trust, Stepping Hill Hospital, Poplar Grove, Hazel Grove, Stockport, SK2 7JE, UK
| | - Stuart Russell
- East Cheshire NHS Trust, Victoria Road, Macclesfield, SK10 3BL, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paula R Williamson
- Department of Biostatistics, University of Liverpool, Member of Liverpool Health Partners, Block F, Waterhouse Bld, 1-5 Brownlow Street, Liverpool, L69 3GL, UK
| | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
- Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK.
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
50
|
Khan M, Meuth AI, Brown SM, Chandrasekar B, Bowles DK, Bender SB. Aldosterone impairs coronary adenosine-mediated vasodilation via reduced functional expression of Ca 2+-activated K + channels. Am J Physiol Heart Circ Physiol 2019; 317:H357-H363. [PMID: 31199187 DOI: 10.1152/ajpheart.00081.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Elevated plasma aldosterone (Aldo) levels are associated with greater risk of cardiac ischemic events and cardiovascular mortality. Adenosine-mediated coronary vasodilation is a critical cardioprotective mechanism during ischemia; however, whether this response is impaired by increased Aldo is unclear. We hypothesized that chronic Aldo impairs coronary adenosine-mediated vasodilation via downregulation of vascular K+ channels. Male C57BL/6J mice were treated with vehicle (Con) or subpressor Aldo for 4 wk. Coronary artery function, assessed by wire myography, revealed Aldo-induced reductions in vasodilation to adenosine and the endothelium-dependent vasodilator acetylcholine but not to the nitric oxide donor sodium nitroprusside. Coronary vasoconstriction to endothelin-1 and the thromboxane A2 mimetic U-46619 was unchanged by Aldo. Additional mechanistic studies revealed impaired adenosine A2A, not A2B, receptor-dependent vasodilation by Aldo with a tendency for Aldo-induced reduction of coronary A2A gene expression. Adenylate cyclase inhibition attenuated coronary adenosine dilation but did not eliminate group differences, and adenosine-stimulated vascular cAMP production was similar between Con and Aldo mice. Similarly, blockade of inward rectifier K+ channels reduced but did not eliminate group differences in adenosine dilation whereas group differences were eliminated by blockade of Ca2+-activated K+ (KCa) channels that blunted and abrogated adenosine and A2A-dependent dilation, respectively. Gene expression of several coronary KCa channels was reduced by Aldo. Together, these data demonstrate Aldo-induced impairment of adenosine-mediated coronary vasodilation involving blunted A2A-KCa-dependent vasodilation, independent of blood pressure, providing important insights into the link between plasma Aldo and cardiac mortality and rationale for aldosterone antagonist use to preserve coronary microvascular function.NEW & NOTEWORTHY Increased plasma aldosterone levels are associated with worsened cardiac outcomes in diverse patient groups by unclear mechanisms. We identified that, in male mice, elevated aldosterone impairs coronary adenosine-mediated vasodilation, an important cardioprotective mechanism. This aldosterone-induced impairment involves reduced adenosine A2A, not A2B, receptor-dependent vasodilation associated with downregulation of coronary KCa channels and does not involve altered adenylate cyclase/cAMP signaling. Importantly, this effect of aldosterone occurred independent of changes in coronary vasoconstrictor responsiveness and blood pressure.
Collapse
Affiliation(s)
- Maloree Khan
- Biomedical Sciences, University of Missouri, Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Alex I Meuth
- Biomedical Sciences, University of Missouri, Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Scott M Brown
- Biomedical Sciences, University of Missouri, Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Bysani Chandrasekar
- Medicine-Cardiology, University of Missouri School of Medicine, Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| | - Douglas K Bowles
- Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri.,Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri
| |
Collapse
|