1
|
Chang YC, Wu XM, Chen TY, Chen UL, Liao CW, Lai TS, Chang CC, Lee BC, Yang FY, Chen ZW, Chang YY, Chueh JS, Wu VC, Tsai CH, Hung CS, Lin YH. Evaluating the effects of adrenalectomy and mineralocorticoid receptor antagonist on cardiac remodeling and diastolic function in patients with aldosterone-producing adenoma. Hypertens Res 2025; 48:529-539. [PMID: 39448809 DOI: 10.1038/s41440-024-01946-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024]
Abstract
Cardiac remodeling and diastolic dysfunction in patients with aldosterone-producing adenomas (APA) can be improved after adrenalectomy. However, the effect of mineralocorticoid receptor antagonist (MRA) treatment remains unclear. The aim of this study is to evaluate the effect of MRA on cardiac remodeling and diastolic dysfunction in patients with PA. We prospectively enrolled patients with APA from 1993 to 2023, who either received medical treatment with MRAs or underwent adrenalectomy. Biochemical characteristics and echocardiographic findings were collected at baseline and one year after treatment. Propensity score matching was conducted based on baseline biochemical characteristics, left ventricular mass index (LVMI), and diastolic function. A total of 467 APA patients were enrolled in the study. After propensity score matching, 159 patients who underwent adrenalectomy were matched with 159 patients who received MRAs. After therapy, patients who received MRAs showed significant improvement in diastolic function after one year of treatment but not LVMI. Compared to the MRA group, the adrenalectomy group had greater improvement in systolic blood pressure, plasma aldosterone concentration, plasma renin activity, aldosterone-to-renin ratio, and LVMI. In multivariable regression analysis, pretreatment echocardiographic values were significantly associated with changes in both LVMI and E/e', while the treatment strategy showed a significant association with changes in LVMI. Thus, one year after therapy, both adrenalectomy and MRA are effective in improving diastolic function in patients with APA. However, adrenalectomy is more effective than MRA treatment in reversing cardiac remodeling in patients with APA.
Collapse
Affiliation(s)
- Yu-Ching Chang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Xue-Ming Wu
- Department of Internal Medicine, Taoyuan General Hospital, Taoyuan, Taiwan
| | - Tsung-Yan Chen
- Division of Cardiology, Department of Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Uei-Lin Chen
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taoyuan Min Sheng General Hospital, Taoyuan, Taiwan
| | - Che-Wei Liao
- Department of Internal Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Tai-Shuan Lai
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chin-Chen Chang
- Department of Medical Imaging, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Bo-Ching Lee
- Department of Medical Imaging, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Fang-Yu Yang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Zheng-Wei Chen
- Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Yi-Yao Chang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Jeff S Chueh
- Department of Urology, National Taiwan University Hospital, Taipei, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Cheng-Hsuan Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chi-Sheng Hung
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan.
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
2
|
Li Z, Williams H, Jackson ML, Johnson JL, George SJ. WISP-1 Regulates Cardiac Fibrosis by Promoting Cardiac Fibroblasts' Activation and Collagen Processing. Cells 2024; 13:989. [PMID: 38891121 PMCID: PMC11172092 DOI: 10.3390/cells13110989] [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: 04/12/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Hypertension induces cardiac fibrotic remodelling characterised by the phenotypic switching of cardiac fibroblasts (CFs) and collagen deposition. We tested the hypothesis that Wnt1-inducible signalling pathway protein-1 (WISP-1) promotes CFs' phenotypic switch, type I collagen synthesis, and in vivo fibrotic remodelling. The treatment of human CFs (HCFs, n = 16) with WISP-1 (500 ng/mL) induced a phenotypic switch (α-smooth muscle actin-positive) and type I procollagen cleavage to an intermediate form of collagen (pC-collagen) in conditioned media after 24h, facilitating collagen maturation. WISP-1-induced collagen processing was mediated by Akt phosphorylation via integrin β1, and disintegrin and metalloproteinase with thrombospondin motifs 2 (ADAMTS-2). WISP-1 wild-type (WISP-1+/+) mice and WISP-1 knockout (WISP-1-/-) mice (n = 5-7) were subcutaneously infused with angiotensin II (AngII, 1000 ng/kg/min) for 28 days. Immunohistochemistry revealed the deletion of WISP-1 attenuated type I collagen deposition in the coronary artery perivascular area compared to WISP-1+/+ mice after a 28-day AngII infusion, and therefore, the deletion of WISP-1 attenuated AngII-induced cardiac fibrosis in vivo. Collectively, our findings demonstrated WISP-1 is a critical mediator in cardiac fibrotic remodelling, by promoting CFs' activation via the integrin β1-Akt signalling pathway, and induced collagen processing and maturation via ADAMTS-2. Thereby, the modulation of WISP-1 levels could provide potential therapeutic targets in clinical treatment.
Collapse
Affiliation(s)
- Ze Li
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Helen Williams
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Molly L. Jackson
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Jason L. Johnson
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
| | - Sarah J. George
- Translational Health Sciences, Bristol Medical School, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK; (Z.L.); (H.W.); (M.L.J.); (J.L.J.)
- Bristol Heart Institute, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Upper Maudlin St, Bristol BS2 8HW, UK
| |
Collapse
|
3
|
Piko N, Bevc S, Hojs R, Ekart R. Finerenone: From the Mechanism of Action to Clinical Use in Kidney Disease. Pharmaceuticals (Basel) 2024; 17:418. [PMID: 38675379 PMCID: PMC11054947 DOI: 10.3390/ph17040418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic kidney disease is a frequent microvascular complication of diabetes and is currently the leading cause of chronic kidney disease and end-stage kidney disease worldwide. Although the prevalence of other complications of diabetes is falling, the number of diabetic patients with end-stage kidney disease in need of kidney replacement therapy is rising. In addition, these patients have extremely high cardiovascular risk. It is more than evident that there is a high unmet treatment need in patients with diabetic kidney disease. Finerenone is a novel nonsteroidal mineralocorticoid receptor antagonist used for treating diabetic kidney disease. It has predominant anti-fibrotic and anti-inflammatory effects and exhibits several renal and cardiac protective effects. This review article summarizes the current knowledge and future prospects of finerenone in treating patients with kidney disease.
Collapse
Affiliation(s)
- Nejc Piko
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia;
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia; (S.B.); (R.H.)
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia; (S.B.); (R.H.)
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, 2000 Maribor, Slovenia;
- Medical Faculty, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
4
|
Ye Z, Okamoto R, Ito H, Ito R, Moriwaki K, Ichikawa M, Kimena L, Ali Y, Ito M, Gomez‐Sanchez CE, Dohi K. Myosin Light Chain Phosphatase Plays an Important Role in Cardiac Fibrosis in a Model of Mineralocorticoid Receptor-Associated Hypertension. J Am Heart Assoc 2024; 13:e032828. [PMID: 38420846 PMCID: PMC10944028 DOI: 10.1161/jaha.123.032828] [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: 09/26/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Myosin phosphatase targeting subunit 2 (MYPT2) is an important subunit of cardiac MLC (myosin light chain) phosphatase, which plays a crucial role in regulating the phosphorylation of MLC to phospho-MLC (p-MLC). A recent study demonstrated mineralocorticoid receptor-related hypertension is associated with RhoA/Rho-associated kinase/MYPT1 signaling upregulation in smooth muscle cells. Our purpose is to investigate the effect of MYPT2 on cardiac function and fibrosis in mineralocorticoid receptor-related hypertension. METHODS AND RESULTS HL-1 murine cardiomyocytes were incubated with different concentrations or durations of aldosterone. After 24-hour stimulation, aldosterone increased CTGF (connective tissue growth factor) and MYPT2 and decreased p-MLC in a dose-dependent manner. MYPT2 knockdown decreased CTGF. Cardiac-specific MYPT2-knockout (c-MYPT2-/-) mice exhibited decreased type 1 phosphatase catalytic subunit β and increased p-MLC. A disease model of mouse was induced by subcutaneous aldosterone and 8% NaCl food for 4 weeks after uninephrectomy. Blood pressure elevation and left ventricular hypertrophy were observed in both c-MYPT2-/- and MYPT2+/+ mice, with no difference in heart weights or nuclear localization of mineralocorticoid receptor in cardiomyocytes. However, c-MYPT2-/- mice had higher ejection fraction and fractional shortening on echocardiography after aldosterone treatment. Histopathology revealed less fibrosis, reduced CTGF, and increased p-MLC in c-MYPT2-/- mice. Basal global radial strain and global longitudinal strain were higher in c-MYPT2-/- than in MYPT2+/+ mice. After aldosterone treatment, both global radial strain and global longitudinal strain remained higher in c-MYPT2-/- mice compared with MYPT2+/+ mice. CONCLUSIONS Cardiac-specific MYPT2 knockout leads to decreased myosin light chain phosphatase and increased p-MLC. MYPT2 deletion prevented cardiac fibrosis and dysfunction in a model of mineralocorticoid receptor-associated hypertension.
Collapse
Affiliation(s)
- Zhe Ye
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| | - Ryuji Okamoto
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
- Regional Medical Support CenterMie University HospitalTsuMieJapan
- Department of Clinical Training and Career Support CenterMie University HospitalTsuMieJapan
| | - Hiromasa Ito
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| | - Rie Ito
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| | - Keishi Moriwaki
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| | - Mizuki Ichikawa
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| | - Lupiya Kimena
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| | - Yusuf Ali
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Masaaki Ito
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| | - Celso E. Gomez‐Sanchez
- Department of Pharmacology and ToxicologyUniversity of Mississippi Medical CenterJacksonMS
| | - Kaoru Dohi
- Department of Cardiology and NephrologyMie University Graduate School of MedicineTsuMieJapan
| |
Collapse
|
5
|
González A, López B, Ravassa S, San José G, Latasa I, Butler J, Díez J. Myocardial Interstitial Fibrosis in Hypertensive Heart Disease: From Mechanisms to Clinical Management. Hypertension 2024; 81:218-228. [PMID: 38084597 DOI: 10.1161/hypertensionaha.123.21708] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Hypertensive heart disease (HHD) can no longer be considered as the beneficial adaptive result of the hypertrophy of cardiomyocytes in response to pressure overload leading to the development of left ventricular hypertrophy. The current evidence indicates that in patients with HHD, pathological lesions in the myocardium lead to maladaptive structural remodeling and subsequent alterations in cardiac function, electrical activity, and perfusion, all contributing to poor outcomes. Diffuse myocardial interstitial fibrosis is probably the most critically involved lesion in these disorders. Therefore, in this review, we will focus on the histological characteristics, the mechanisms, and the clinical consequences of myocardial interstitial fibrosis in patients with HHD. In addition, we will consider the most useful tools for the noninvasive diagnosis of myocardial interstitial fibrosis in patients with HHD, as well as the most effective available therapeutic strategies to prevent its development or facilitate its regression in this patient population. Finally, we will issue a call to action for the need for more fundamental and clinical research on myocardial interstitial fibrosis in HHD.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Department of Pathology, Anatomy and Physiology, Universidad de Navarra, Pamplona, Spain (A.G.)
| | - Begoña López
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Susana Ravassa
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Gorka San José
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Iñigo Latasa
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX (J.B.)
- Department of Medicine, University of Mississippi, Jackson (J.B.)
| | - Javier Díez
- Program of Cardiovascular Disease, Centro de Investigación Médica Aplicada Universidad de Navarra (CIMA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Insitituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
- Center for Biomedical Research in Cardiovascular Diseases Network (CIBERCV), Carlos III Institute of Health, Madrid, Spain (A.G., B.L., S.R., G.S.J., I.L., J.D.)
| |
Collapse
|
6
|
Kodsi M, Makarious D, Gan GC, Choudhary P, Thomas L. Cardiac reverse remodelling by imaging parameters with recent changes to guideline medical therapy in heart failure. ESC Heart Fail 2023; 10:3258-3275. [PMID: 37871982 PMCID: PMC10682888 DOI: 10.1002/ehf2.14555] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/29/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023] Open
Abstract
Recently established heart failure therapies, including sodium glucose co-transporter 2 inhibitors, angiotensin-neprilysin inhibitors, and cardiac resynchronization therapy, have led to both clinical and structural improvements. Reverse remodelling describes the structural and functional responses to therapy and has been shown to correlate with patients' clinical response, acting as a biomarker for treatment success. The introduction of these new therapeutic agents in addition to advances in non-invasive cardiac imaging has led to an expansion in the evaluation and the validation of cardiac reverse remodelling. Methods including volumetric changes as well as strain and myocardial work have all been shown to be non-invasive end-points of reverse remodelling, correlating with clinical outcomes. Our review summarizes the current available evidence on reverse remodelling in heart failure by the non-invasive cardiac imaging techniques, in particular transthoracic echocardiography.
Collapse
Affiliation(s)
- Matthew Kodsi
- Department of CardiologyWestmead HospitalSydneyAustralia
| | - David Makarious
- Department of CardiologyWestmead HospitalSydneyAustralia
- Westmead Clinical School, Westmead HospitalUniversity of SydneySydneyAustralia
| | - Gary C.H. Gan
- Department of CardiologyWestmead HospitalSydneyAustralia
| | - Preeti Choudhary
- Department of CardiologyWestmead HospitalSydneyAustralia
- Westmead Clinical School, Westmead HospitalUniversity of SydneySydneyAustralia
| | - Liza Thomas
- Department of CardiologyWestmead HospitalSydneyAustralia
- Westmead Clinical School, Westmead HospitalUniversity of SydneySydneyAustralia
- South West clinical SchoolUniversity of New South WalesSydneyAustralia
| |
Collapse
|
7
|
Kiyomoto K, Matsuo I, Suita K, Ohnuki Y, Ishikawa M, Ito A, Mototani Y, Tsunoda M, Morii A, Nariyama M, Hayakawa Y, Amitani Y, Gomi K, Okumura S. Oral angiotensin-converting enzyme inhibitor captopril protects the heart from Porphyromonas gingivalis LPS-induced cardiac dysfunction in mice. PLoS One 2023; 18:e0292624. [PMID: 37983238 PMCID: PMC10659197 DOI: 10.1371/journal.pone.0292624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 09/25/2023] [Indexed: 11/22/2023] Open
Abstract
Although angiotensin converting enzyme (ACE) inhibitors are considered useful for the treatment of human heart failure, some experimental failing-heart models have shown little beneficial effect of ACE inhibitors in animals with poor oral health, particularly periodontitis. In this study, we examined the effects of the ACE inhibitor captopril (Cap; 0.1 mg/mL in drinking water) on cardiac dysfunction in mice treated with Porphyromonas gingivalis lipopolysaccharide (PG-LPS) at a dose (0.8 mg/kg/day) equivalent to the circulating level in patients with periodontal disease. Mice were divided into four groups: 1) Control, 2) PG-LPS, 3) Cap, and 4) PG-LPS + Cap. After1 week, we evaluated cardiac function by echocardiography. The left ventricular ejection fraction was significantly decreased in PG-LPS-treated mice compared to the control (from 66 ± 1.8 to 59 ± 2.5%), while Cap ameliorated the dysfunction (63 ± 1.1%). The area of cardiac fibrosis was significantly increased (approximately 2.9-fold) and the number of apoptotic myocytes was significantly increased (approximately 5.6-fold) in the heart of PG-LPS-treated group versus the control, and these changes were suppressed by Cap. The impairment of cardiac function in PG-LPS-treated mice was associated with protein kinase C δ phosphorylation (Tyr-311), leading to upregulation of NADPH oxidase 4 and xanthine oxidase, and calmodulin kinase II phosphorylation (Thr-286) with increased phospholamban phosphorylation (Thr-17). These changes were also suppressed by Cap. Our results suggest that the renin-angiotensin system might play an important role in the development of cardiac diseases induced by PG-LPS.
Collapse
Affiliation(s)
- Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasuharu Amitani
- Department of Mathematics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
8
|
Naaman SC, Bakris GL. Diabetic Nephropathy: Update on Pillars of Therapy Slowing Progression. Diabetes Care 2023; 46:1574-1586. [PMID: 37625003 PMCID: PMC10547606 DOI: 10.2337/dci23-0030] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/16/2023] [Indexed: 08/27/2023]
Abstract
Management of diabetic kidney disease (DKD) has evolved in parallel with our growing understanding of the multiple interrelated pathophysiological mechanisms that involve hemodynamic, metabolic, and inflammatory pathways. These pathways and others play a vital role in the initiation and progression of DKD. Since its initial discovery, the blockade of the renin-angiotensin system has remained a cornerstone of DKD management, leaving a large component of residual risk to be dealt with. The advent of sodium-glucose cotransporter 2 inhibitors followed by nonsteroidal mineralocorticoid receptor antagonists and, to some extent, glucagon-like peptide 1 receptor agonists (GLP-1 RAs) has ushered in a resounding paradigm shift that supports a pillared approach in maximizing treatment to reduce outcomes. This pillared approach is like that derived from the approach to heart failure treatment. The approach mandates that all agents that have been shown in clinical trials to reduce cardiovascular outcomes and/or mortality to a greater extent than a single drug class alone should be used in combination. In this way, each drug class focuses on a specific aspect of the disease's pathophysiology. Thus, in heart failure, β-blockers, sacubitril/valsartan, a mineralocorticoid receptor antagonist, and a diuretic are used together. In this article, we review the evolution of the pillar concept of therapy as it applies to DKD and discuss how it should be used based on the outcome evidence. We also discuss the exciting possibility that GLP-1 RAs may be an additional pillar in the quest to further slow kidney disease progression in diabetes.
Collapse
Affiliation(s)
- Sandra C. Naaman
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and American Heart Association Comprehensive Hypertension Center, University of Chicago Medicine, Chicago, IL
| | - George L. Bakris
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and American Heart Association Comprehensive Hypertension Center, University of Chicago Medicine, Chicago, IL
| |
Collapse
|
9
|
Smart CD, Madhur MS. The immunology of heart failure with preserved ejection fraction. Clin Sci (Lond) 2023; 137:1225-1247. [PMID: 37606086 PMCID: PMC10959189 DOI: 10.1042/cs20230226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/23/2023] [Accepted: 07/31/2023] [Indexed: 08/23/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) now accounts for the majority of new heart failure diagnoses and continues to increase in prevalence in the United States. Importantly, HFpEF is a highly morbid, heterogeneous syndrome lacking effective therapies. Inflammation has emerged as a potential contributor to the pathogenesis of HFpEF. Many of the risk factors for HFpEF are also associated with chronic inflammation, such as obesity, hypertension, aging, and renal dysfunction. A large amount of preclinical evidence suggests that immune cells and their associated cytokines play important roles in mediating fibrosis, oxidative stress, metabolic derangements, and endothelial dysfunction, all potentially important processes in HFpEF. How inflammation contributes to HFpEF pathogenesis, however, remains poorly understood. Recently, a variety of preclinical models have emerged which may yield much needed insights into the causal relationships between risk factors and the development of HFpEF, including the role of specific immune cell subsets or inflammatory pathways. Here, we review evidence in animal models and humans implicating inflammation as a mediator of HFpEF and identify gaps in knowledge requiring further study. As the understanding between inflammation and HFpEF evolves, it is hoped that a better understanding of the mechanisms underlying immune cell activation in HFpEF can open up new therapeutic avenues.
Collapse
Affiliation(s)
- Charles Duncan Smart
- Department of Molecular Physiology and Biophysics,
Vanderbilt University School of Medicine, Nashville, TN, U.S.A
| | - Meena S. Madhur
- Department of Molecular Physiology and Biophysics,
Vanderbilt University School of Medicine, Nashville, TN, U.S.A
- Department of Medicine, Division of Cardiovascular
Medicine, Vanderbilt University Medical Center, Nashville, TN, U.S.A
- Department of Medicine, Division of Clinical Pharmacology,
Vanderbilt University Medical Center, Nashville, TN, U.S.A
- Vanderbilt Institute for Infection, Immunology, and
Inflammation, Nashville, TN, U.S.A
| |
Collapse
|
10
|
Pillai K, Fares A, Dargham S, Al Suwaidi J, Jayyousi A, Abi Khalil C. Primary hyperaldosteronism is associated with increased mortality and morbidity in patients with hypertension and diabetes. Front Endocrinol (Lausanne) 2023; 14:1147225. [PMID: 37305032 PMCID: PMC10250736 DOI: 10.3389/fendo.2023.1147225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 05/15/2023] [Indexed: 06/13/2023] Open
Abstract
Aims Primary hyperaldosteronism (PA) is a common cause of hypertension. It is more prevalent in patients with diabetes. We assessed the cardiovascular impact of PA in patients with established hypertension and diabetes. Methods Data from the National Inpatient Sample (2008-2016) was used to identify adults with PA with hypertension and diabetes comorbidities and then compared to non-PA patients. The primary outcome was in-hospital death. Secondary outcomes included ischemic stroke, hemorrhagic stroke, acute renal failure, atrial fibrillation, and acute heart failure. Results A total of 48,434,503 patients with hypertension and diabetes were included in the analysis, of whom 12,850 (0.03%) were diagnosed with primary hyperaldosteronism (PA). Compared to patients with hypertension and diabetes but no PA, those with PA were more likely to be younger [63(13) vs. 67 (14), male (57.1% vs. 48.3%), and African-Americans (32% vs. 18.5%) (p<0.001 for all). PA was associated with a higher risk of mortality (adjusted OR 1.076 [1.076-1.077]), ischemic stroke [adjusted OR 1.049 (1.049-1.05)], hemorrhagic stroke [adjusted OR 1.05 (1.05-1.051)], acute renal failure [adjusted OR 1.058 (1.058-1.058)], acute heart failure [OR 1.104 (1.104-1.104)], and atrial fibrillation [adjusted OR 1.034 (1.033-1.034)]. As expected, older age and underlying cardiovascular disease were the strongest predictors of mortality. However, the female gender conferred protection [OR 0.889 (0.886-0.892]. Conclusion Primary hyperaldosteronism in patients with hypertension and diabetes is associated with increased mortality and morbidity.
Collapse
Affiliation(s)
| | - Ahmed Fares
- Department of Medicine, Weill Cornell Medicine–Qatar, Doha, Qatar
| | - Soha Dargham
- Biostatistics Core, Weill Cornell Medicine–Qatar, Doha, Qatar
| | | | - Amin Jayyousi
- Department of Endocrinology, Hamad Medical Corporation, Doha, Qatar
| | - Charbel Abi Khalil
- Department of Medicine, Weill Cornell Medicine–Qatar, Doha, Qatar
- Heart Hospital, Hamad Medical Corporation, Doha, Qatar
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
11
|
Jerome JR, Deliyanti D, Suphapimol V, Kolkhof P, Wilkinson-Berka JL. Finerenone, a Non-Steroidal Mineralocorticoid Receptor Antagonist, Reduces Vascular Injury and Increases Regulatory T-Cells: Studies in Rodents with Diabetic and Neovascular Retinopathy. Int J Mol Sci 2023; 24:ijms24032334. [PMID: 36768656 PMCID: PMC9917037 DOI: 10.3390/ijms24032334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Vision loss in diabetic retinopathy features damage to the blood-retinal barrier and neovascularization, with hypertension and the renin-angiotensin system (RAS) having causal roles. We evaluated if finerenone, a non-steroidal mineralocorticoid receptor (MR) antagonist, reduced vascular pathology and inflammation in diabetic and neovascular retinopathy. Diabetic and hypertensive transgenic (mRen-2)27 rats overexpressing the RAS received the MR antagonist finerenone (10 mg/kg/day, oral gavage) or the angiotensin-converting enzyme inhibitor perindopril (10 mg/kg/day, drinking water) for 12 weeks. As retinal neovascularization does not develop in diabetic rodents, finerenone (5 mg/kg/day, i.p.) was evaluated in murine oxygen-induced retinopathy (OIR). Retinal vasculopathy was assessed by measuring gliosis, vascular leakage, neovascularization, and VEGF. Inflammation was investigated by quantitating retinal microglia/macrophages, pro-inflammatory mediators, and anti-inflammatory regulatory T-cells (Tregs). In diabetes, both treatments reduced systolic blood pressure, gliosis, vascular leakage, and microglial/macrophage density, but only finerenone lowered VEGF, ICAM-1, and IL-1ß. In OIR, finerenone reduced neovascularization, vascular leakage, and microglial density, and increased Tregs in the blood, spleen, and retina. Our findings, in the context of the FIDELIO-DKD and FIGARO-DKD trials reporting the benefits of finerenone on renal and cardiovascular outcomes in diabetic kidney disease, indicate the potential of finerenone as an effective oral treatment for diabetic retinopathy.
Collapse
Affiliation(s)
- Jack R. Jerome
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Devy Deliyanti
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Varaporn Suphapimol
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | | | - Jennifer L. Wilkinson-Berka
- Department of Anatomy and Physiology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
- Correspondence:
| |
Collapse
|
12
|
Cardiovascular Disease in Obstructive Sleep Apnea: Putative Contributions of Mineralocorticoid Receptors. Int J Mol Sci 2023; 24:ijms24032245. [PMID: 36768567 PMCID: PMC9916750 DOI: 10.3390/ijms24032245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic and highly prevalent condition that is associated with oxidative stress, inflammation, and fibrosis, leading to endothelial dysfunction, arterial stiffness, and vascular insulin resistance, resulting in increased cardiovascular disease and overall mortality rates. To date, OSA remains vastly underdiagnosed and undertreated, with conventional treatments yielding relatively discouraging results for improving cardiovascular outcomes in OSA patients. As such, a better mechanistic understanding of OSA-associated cardiovascular disease (CVD) and the development of novel adjuvant therapeutic targets are critically needed. It is well-established that inappropriate mineralocorticoid receptor (MR) activation in cardiovascular tissues plays a causal role in a multitude of CVD states. Clinical studies and experimental models of OSA lead to increased secretion of the MR ligand aldosterone and excessive MR activation. Furthermore, MR activation has been associated with worsened OSA prognosis. Despite these documented relationships, there have been no studies exploring the causal involvement of MR signaling in OSA-associated CVD. Further, scarce clinical studies have exclusively assessed the beneficial role of MR antagonists for the treatment of systemic hypertension commonly associated with OSA. Here, we provide a comprehensive overview of overlapping mechanistic pathways recruited in the context of MR activation- and OSA-induced CVD and propose MR-targeted therapy as a potential avenue to abrogate the deleterious cardiovascular consequences of OSA.
Collapse
|
13
|
Chang YY, Lee BC, Chen ZW, Tsai CH, Chang CC, Liao CW, Pan CT, Peng KY, Chou CH, Lu CC, Wu VC, Hung CS, Lin YH, TAIPAI study group. Cardiovascular and metabolic characters of KCNJ5 somatic mutations in primary aldosteronism. Front Endocrinol (Lausanne) 2023; 14:1061704. [PMID: 36950676 PMCID: PMC10025475 DOI: 10.3389/fendo.2023.1061704] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Primary aldosteronism (PA) is the leading cause of curable endocrine hypertension, which is associated with a higher risk of cardiovascular and metabolic insults compared to essential hypertension. Aldosterone-producing adenoma (APA) is a major cause of PA, which can be treated with adrenalectomy. Somatic mutations are the main pathogenesis of aldosterone overproduction in APA, of which KCNJ5 somatic mutations are most common, especially in Asian countries. This article aimed to review the literature on the impacts of KCNJ5 somatic mutations on systemic organ damage. EVIDENCE ACQUISITION PubMed literature research using keywords combination, including "aldosterone-producing adenoma," "somatic mutations," "KCNJ5," "organ damage," "cardiovascular," "diastolic function," "metabolic syndrome," "autonomous cortisol secretion," etc. RESULTS APA patients with KCNJ5 somatic mutations are generally younger, female, have higher aldosterone levels, lower potassium levels, larger tumor size, and higher hypertension cure rate after adrenalectomy. This review focuses on the cardiovascular and metabolic aspects of KCNJ5 somatic mutations in APA patients, including left ventricular remodeling and diastolic function, abdominal aortic thickness and calcification, arterial stiffness, metabolic syndrome, abdominal adipose tissue, and correlation with autonomous cortisol secretion. Furthermore, we discuss modalities to differentiate the types of mutations before surgery. CONCLUSION KCNJ5 somatic mutations in patients with APA had higher left ventricular mass (LVM), more impaired diastolic function, thicker aortic wall, lower incidence of metabolic syndrome, and possibly a lower incidence of concurrent autonomous cortisol secretion, but better improvement in LVM, diastolic function, arterial stiffness, and aortic wall thickness after adrenalectomy compared to patients without KCNJ5 mutations.
Collapse
Affiliation(s)
- Yi-Yao Chang
- Cardiology Division of Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
| | - Bo-Ching Lee
- Department of Medical Imaging, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Zheng-Wei Chen
- Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Cheng-Hsuan Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chin-Chen Chang
- Department of Medical Imaging, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Che-Wei Liao
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Chien-Ting Pan
- Department of Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Kang-Yung Peng
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Hung Chou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Chu Lu
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Vin-Cent Wu
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Sheng Hung
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan
- *Correspondence: Yen-Hung Lin,
| | | |
Collapse
|
14
|
Abstract
Besides the physiological regulation of water, sodium, and potassium homeostasis, aldosterone modulates several physiological and pathological processes in the cardiovascular system. At the vascular level, aldosterone excess stimulates endothelial dysfunction and infiltration of inflammatory cells, enhances the development of the atherosclerotic plaque, and favors plaque instability, arterial stiffness, and calcification. At the cardiac level, aldosterone increases cardiac inflammation, fibrosis, and myocardial hypertrophy. As a clinical consequence, high aldosterone levels are associated with enhanced risk of cardiovascular events and mortality, especially when aldosterone secretion is inappropriate for renin levels and sodium intake, as in primary aldosteronism. Several clinical trials showed that mineralocorticoid receptor antagonists reduce cardiovascular mortality in patients with heart failure and reduced ejection fraction, but inconclusive results were reported for other cardiovascular conditions, such as heart failure with preserved ejection fraction, myocardial infarction, and atrial fibrillation. In patients with primary aldosteronism, adrenalectomy or treatment with mineralocorticoid receptor antagonists significantly mitigate adverse aldosterone effects, reducing the risk of cardiovascular events, mortality, and incident atrial fibrillation. In this review, we will summarize the major preclinical and clinical studies investigating the cardiovascular damage mediated by aldosterone and the protective effect of mineralocorticoid receptor antagonists for the reduction of cardiovascular risk in patients with cardiovascular diseases and primary aldosteronism.
Collapse
Affiliation(s)
- Fabrizio Buffolo
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Martina Tetti
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Paolo Mulatero
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| | - Silvia Monticone
- Division of Internal Medicine and Hypertension Unit, Department of Medical Sciences, University of Torino, Italy
| |
Collapse
|
15
|
Travers JG, Tharp CA, Rubino M, McKinsey TA. Therapeutic targets for cardiac fibrosis: from old school to next-gen. J Clin Invest 2022; 132:148554. [PMID: 35229727 PMCID: PMC8884906 DOI: 10.1172/jci148554] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases remain the leading cause of death worldwide, with pathological fibrotic remodeling mediated by activated cardiac myofibroblasts representing a unifying theme across etiologies. Despite the profound contributions of myocardial fibrosis to cardiac dysfunction and heart failure, there currently exist limited clinical interventions that effectively target the cardiac fibroblast and its role in fibrotic tissue deposition. Exploration of novel strategies designed to mitigate or reverse myofibroblast activation and cardiac fibrosis will likely yield powerful therapeutic approaches for the treatment of multiple diseases of the heart, including heart failure with preserved or reduced ejection fraction, acute coronary syndrome, and cardiovascular disease linked to type 2 diabetes. In this Review, we provide an overview of classical regulators of cardiac fibrosis and highlight emerging, next-generation epigenetic regulatory targets that have the potential to revolutionize treatment of the expanding cardiovascular disease patient population.
Collapse
|
16
|
Clinical Pharmacokinetics and Pharmacodynamics of Esaxerenone, a Novel Mineralocorticoid Receptor Antagonist: A Review. Eur J Drug Metab Pharmacokinet 2022; 47:291-308. [PMID: 35190999 DOI: 10.1007/s13318-022-00760-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2022] [Indexed: 12/11/2022]
Abstract
Esaxerenone is a selective, nonsteroidal, high-affinity mineralocorticoid receptor antagonist recently approved in Japan for the treatment of hypertension. It has high oral biovailability, a large volume of distribution, and is primarly metabolized in liver and excreted in bile. Esaxerenone is an efficient antihypertensive, whether given alone or as add-on therapy. The antihypertensive effect is accompanied by renoprotective action, which is being further investigated in current clinical trials. Due to its relatively long half-life and high affinity for the mineralocorticoid receptor, esaxerenone is administered once daily and in low absolute doses. The safety of esaxerenone is considerable, since hyperkalemia is not frequent and, when it does appear, not sustained. Endocrine adverse events, which frequently occur with steroidal mineralocorticoid receptor antagonists, are extremely rare with esaxerenone. Although the risk of clinically significant drug-drug interactions is not high, esaxerenone treatment should start with low doses, with subsequent titration to achieve the optimal clinical effect, all while monitoring serum potassium and paying attention to concomitant therapy with drugs that may induce or inhibit esaxerenone metabolism. This review article offers comprehensive information about the pharmacodynamics and pharmacokinetics of esaxerenone in humans, which should help clinicians to more precisely tailor esaxerenone dosing regimens to their patients.
Collapse
|
17
|
Barrera-Chimal J, Lima-Posada I, Bakris GL, Jaisser F. Mineralocorticoid receptor antagonists in diabetic kidney disease - mechanistic and therapeutic effects. Nat Rev Nephrol 2022; 18:56-70. [PMID: 34675379 DOI: 10.1038/s41581-021-00490-8] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 01/19/2023]
Abstract
Chronic kidney disease (CKD) is the leading complication in type 2 diabetes (T2D) and current therapies that limit CKD progression and the development of cardiovascular disease (CVD) include angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and sodium-glucose co-transporter 2 (SGLT2) inhibitors. Despite the introduction of these therapeutics, an important residual risk of CKD progression and cardiovascular death remains in patients with T2D. Mineralocorticoid receptor antagonists (MRAs) are a promising therapeutic option in diabetic kidney disease (DKD) owing to the reported effects of mineralocorticoid receptor activation in inflammatory cells, podocytes, fibroblasts, mesangial cells and vascular cells. In preclinical studies, MRAs consistently reduce albuminuria, CKD progression, and activation of fibrotic and inflammatory pathways. DKD clinical studies have similarly demonstrated that steroidal MRAs lead to albuminuria reduction compared with placebo, although hyperkalaemia is a major secondary effect. Non-steroidal MRAs carry a lower risk of hyperkalaemia than steroidal MRAs, and the large FIDELIO-DKD clinical trial showed that the non-steroidal MRA finerenone also slowed CKD progression and reduced the risk of adverse cardiovascular outcomes compared with placebo in patients with T2D. Encouragingly, other non-steroidal MRAs have anti-albuminuric properties in DKD. Whether or not combining MRAs with other renoprotective drugs such as SGLT2 inhibitors might provide additive protective effects warrants further investigation.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico.,Laboratorio de Fisiología Cardiovascular y Trasplante Renal, Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Ixchel Lima-Posada
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France
| | - George L Bakris
- American Heart Association Comprehensive Hypertension Centre, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Frederic Jaisser
- INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, France. .,Université de Lorraine, INSERM Centre d'Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France.
| |
Collapse
|
18
|
de Jesus ÉF, Nunes ADDC, Pontes CNR, Macedo LM, Mendes EP, Ianzer DA, da Costa M, Ghedini PC, Dos Santos FCA, Biancardi MF, Castro CH. Cardioprotective effects of the proline-rich oligopeptide Bj-PRO-7a in spontaneously hypertensive rats. Clin Exp Pharmacol Physiol 2021; 48:1693-1703. [PMID: 34427931 DOI: 10.1111/1440-1681.13577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 08/03/2021] [Accepted: 08/20/2021] [Indexed: 11/28/2022]
Abstract
The proline-rich oligopeptide from Bothrops jararaca snake venom, Bj-PRO-7a, promotes acute effects in blood pressure in hypertensive animals. However, the cardiac effects of this heptapeptide are completely unknown. Thus, we sought to evaluate whether the Bj-PRO-7a could protect against cardiac remodelling in spontaneously hypertensive rats (SHR). SHR were treated with Bj-PRO-7a (71 nmol/kg/day, s.c.) or saline for 28 days. Wistar rats were used as control. Systolic blood pressure (SBP) and heart rate (HR) were measured by tail-cuff plethysmography. Cardiomyocyte diameter and interstitial and perivascular fibrosis of the left ventricle (LV) were evaluated using Picrosirius staining. Immunofluorescence was used to detect collagen I and III. Fibroblast proliferation was assessed by immunohistochemistry to detect proliferating cell nuclear antigen (PCNA). Protein expression was assessed by western blot. The superoxide dismutase and catalase activities and the concentration of lipid peroxidation products were evaluated in the LV. The SBP and HR were not different between treated and non-treated SHR at the end of the treatment. However, Bj-PRO-7a attenuated the cardiomyocyte hypertrophy, deposition of interstitial and perivascular fibrosis and collagen I, and positive PCNA-labelled fibroblasts. This peptide also reduced the increased levels of TBARS, expression and activity of catalase, and activity of SOD in LV from SHR. Also, the Bj-PRO-7a increased the expression of metalloproteinases-2 in SHR hearts. These findings demonstrate that the Bj-PRO-7a reduced the pathological cardiac remodelling in a pressure-independent manner in hypertensive rats through mechanisms mediated by oxidative stress regulation.
Collapse
Affiliation(s)
- Érika Fernandes de Jesus
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Allancer Divino de Carvalho Nunes
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Carolina Nobre Ribeiro Pontes
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Larissa Matuda Macedo
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Elizabeth Pereira Mendes
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Danielle Alves Ianzer
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Michael da Costa
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Paulo César Ghedini
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | - Manoel Francisco Biancardi
- Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Carlos Henrique Castro
- Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| |
Collapse
|
19
|
Epstein M, Freundlich M. The intersection of Mineralocorticoid Receptor (MR) activation and the FGF23 - Klotho cascade. A Duopoly that promotes renal and cardiovascular injury. Nephrol Dial Transplant 2021; 37:211-221. [PMID: 34459924 DOI: 10.1093/ndt/gfab254] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
The nexus of CKD and cardiovascular disease (CVD) amplifies the morbidity and mortality of CKD, emphasizing the need for defining and establishing therapeutic initiatives to modify and abrogate the progression of CKD and concomitant CV risks. In addition to the traditional CV risk factors, disturbances of mineral metabolism are specific risk factors that contribute to the excessive CV mortality in patients with CKD. These risk factors include dysregulations of circulating factors that modulate phosphate metabolism including fibroblast growth factor 23 (FGF23) and soluble Klotho. Reduced circulating levels and suppressed renal klotho expression may be associated with adverse outcomes in CKD patients. While elevated circulating concentrations or locally produced FGF23 in the strained heart exert pro-hypertrophic mechanisms on the myocardium, Klotho attenuates tissue fibrosis, progression of CKD, cardiomyopathy, endothelial dysfunction, vascular stiffness, and vascular calcification. Mineralocorticoid receptor (MR) activation in non-classical targets, mediated by aldosterone and other ligands, amplifies CVD in CKD. In concert, we detail how the interplay of elevated FGF23, activation of the MR, and concomitant reductions of circulating Klotho in CKD, may potentiate each other's deleterious effects on kidney and the heart, thereby contributing to the initiation and progression of kidney and cardiac functional deterioration, acting through multipronged albeit complementary mechanistic pathways.
Collapse
Affiliation(s)
- Murray Epstein
- Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael Freundlich
- Division of Pediatric Nephrology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
20
|
Burrello J, Tetti M, Forestiero V, Biemmi V, Bolis S, Pomatto MAC, Amongero M, Di Silvestre D, Mauri P, Vassalli G, Camussi G, Williams TA, Mulatero P, Barile L, Monticone S. Characterization of Circulating Extracellular Vesicle Surface Antigens in Patients With Primary Aldosteronism. Hypertension 2021; 78:726-737. [PMID: 34304584 DOI: 10.1161/hypertensionaha.121.17136] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jacopo Burrello
- From the Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Turin, Italy (J.B., M.T., V.F., T.A.W., P. Mulatero, S.M.).,Laboratory for Cardiovascular Theranostics and Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, Lugano, Switzerland (J.B., V.B., S.B., G.V., L.B.)
| | - Martina Tetti
- From the Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Turin, Italy (J.B., M.T., V.F., T.A.W., P. Mulatero, S.M.)
| | - Vittorio Forestiero
- From the Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Turin, Italy (J.B., M.T., V.F., T.A.W., P. Mulatero, S.M.)
| | - Vanessa Biemmi
- Laboratory for Cardiovascular Theranostics and Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, Lugano, Switzerland (J.B., V.B., S.B., G.V., L.B.).,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland (V.B., L.B.)
| | - Sara Bolis
- Laboratory for Cardiovascular Theranostics and Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, Lugano, Switzerland (J.B., V.B., S.B., G.V., L.B.)
| | | | - Martina Amongero
- Department of Mathematical Sciences G. L. Lagrange, Polytechnic University of Torino, Italy (M.A.)
| | - Dario Di Silvestre
- Proteomic and Metabolomic Laboratory, Institute for Biomedical Technologies-National Research Council (ITB-CNR), Segrate (Milan), Italy (D.D.S., P. Mauri)
| | - Pierluigi Mauri
- Proteomic and Metabolomic Laboratory, Institute for Biomedical Technologies-National Research Council (ITB-CNR), Segrate (Milan), Italy (D.D.S., P. Mauri)
| | - Giuseppe Vassalli
- Laboratory for Cardiovascular Theranostics and Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, Lugano, Switzerland (J.B., V.B., S.B., G.V., L.B.)
| | - Giovanni Camussi
- Molecular Biotechnology Center, Department of Medical Sciences, University of Torino, Italy (M.A.C.P., G.C.)
| | - Tracy Ann Williams
- From the Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Turin, Italy (J.B., M.T., V.F., T.A.W., P. Mulatero, S.M.).,Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany (T.A.W)
| | - Paolo Mulatero
- From the Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Turin, Italy (J.B., M.T., V.F., T.A.W., P. Mulatero, S.M.)
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics and Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, Lugano, Switzerland (J.B., V.B., S.B., G.V., L.B.).,Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland (V.B., L.B.)
| | - Silvia Monticone
- From the Division of Internal Medicine 4 and Hypertension Unit, Department of Medical Sciences, University of Turin, Italy (J.B., M.T., V.F., T.A.W., P. Mulatero, S.M.)
| |
Collapse
|
21
|
Wu T, Ren Y, Wang W, Cheng W, Zhou F, He S, Liu X, Li L, Tang L, Deng Q, Zhou X, Chen Y, Sun J. Left Ventricular Remodeling in Patients with Primary Aldosteronism: A Prospective Cardiac Magnetic Resonance Imaging Study. Korean J Radiol 2021; 22:1619-1627. [PMID: 34269528 PMCID: PMC8484156 DOI: 10.3348/kjr.2020.1291] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/07/2021] [Accepted: 04/25/2021] [Indexed: 02/05/2023] Open
Abstract
Objective This study used cardiac magnetic resonance imaging (MRI) to compare the characteristics of left ventricular remodeling in patients with primary aldosteronism (PA) with those of patients with essential hypertension (EH) and healthy controls (HCs). Materials and Methods This prospective study enrolled 35 patients with PA, in addition to 35 age- and sex-matched patients with EH, and 35 age- and sex-matched HCs, all of whom underwent comprehensive clinical and cardiac MRI examinations. The analysis of variance was used to detect the differences in the characteristics of left ventricular remodeling among the three groups. Univariable and multivariable linear regression analyses were used to determine the relationships between left ventricular remodeling and the physiological variables. Results The left ventricular end-diastolic volume index (EDVi) (mean ± standard deviation [SD]: 85.1 ± 13.0 mL/m2 for PA, 75.9 ± 14.3 mL/m2 for EH, and 77.3 ± 12.8 mL/m2 for HC; p = 0.010), left ventricular end-systolic volume index (ESVi) (mean ± SD: 35.2 ± 9.8 mL/m2 for PA, 30.7 ± 8.1 mL/m2 for EH, and 29.5 ± 7.0 mL/m2 for HC; p = 0.013), left ventricular mass index (mean ± SD: 65.8 ± 16.5 g/m2 for PA, 56.9 ± 12.1 g/m2 for EH, and 44.1 ± 8.9 g/m2 for HC; p < 0.001), and native T1 (mean ± SD: 1224 ± 39 ms for PA, 1201 ± 47 ms for EH, and 1200 ± 44 ms for HC; p = 0.041) values were higher in the PA group compared to the EH and HC groups. Multivariable linear regression demonstrated that log (plasma aldosterone-to-renin ratio) was independently correlated with EDVi and ESVi. Plasma aldosterone was independently correlated with native T1. Conclusion Patients with PA showed a greater degree of ventricular hypertrophy and enlargement, as well as myocardial fibrosis, compared to those with EH. Cardiac MRI T1 mapping can detect left ventricular myocardial fibrosis in patients with PA.
Collapse
Affiliation(s)
- Tao Wu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Ren
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Cheng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Fangli Zhou
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Shuai He
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiumin Liu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Tang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Deng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyue Zhou
- Department of North Sichuan Medical College, Nanchong, China
| | - Yucheng Chen
- Department of MR Collaboration, Siemens Healthcare, Shanghai, China
| | - Jiayu Sun
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
22
|
Vodošek Hojs N, Bevc S, Ekart R, Piko N, Petreski T, Hojs R. Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease. Pharmaceuticals (Basel) 2021; 14:561. [PMID: 34208285 PMCID: PMC8230766 DOI: 10.3390/ph14060561] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Diabetes mellitus is a global health issue and main cause of chronic kidney disease. Both diseases are also linked through high cardiovascular morbidity and mortality. Diabetic kidney disease (DKD) is present in up to 40% of diabetic patients; therefore, prevention and treatment of DKD are of utmost importance. Much research has been dedicated to the optimization of DKD treatment. In the last few years, mineralocorticoid receptor antagonists (MRA) have experienced a renaissance in this field with the development of non-steroidal MRA. Steroidal MRA have known cardiorenal benefits, but their use is limited by side effects, especially hyperkalemia. Non-steroidal MRA still block the damaging effects of mineralocorticoid receptor overactivation (extracellular fluid volume expansion, inflammation, fibrosis), but with fewer side effects (hormonal, hyperkalemia) than steroidal MRA. This review article summarizes the current knowledge and newer research conducted on MRA in DKD.
Collapse
Affiliation(s)
- Nina Vodošek Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
| | - Robert Ekart
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Nejc Piko
- Department of Dialysis, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia;
| | - Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
| | - Radovan Hojs
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000 Maribor, Slovenia; (S.B.); (T.P.); (R.H.)
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia;
| |
Collapse
|
23
|
Epstein M. Aldosterone and Mineralocorticoid Receptor Signaling as Determinants of Cardiovascular and Renal Injury: From Hans Selye to the Present. Am J Nephrol 2021; 52:209-216. [PMID: 33857953 DOI: 10.1159/000515622] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND A full understanding of the mechanisms of action of aldosterone and its interaction with the mineralocorticoid receptor (MR) allows a theoretical framework to predict the therapeutic potential of MR antagonists (MRAs) in CKD, and heart failure with reduced ejection fraction. SUMMARY The initial focus on the mechanisms of action of aldosterone was directed primarily on its role in modulating renal excretory function. In contrast, many recent studies have demonstrated a wider and expanded role for aldosterone in modulating inflammation, collagen formation, fibrosis, and necrosis. Increasing evidence has accrued that implicates the pathophysiological overactivation of the MR as a major determinant of progression of CKD. By promoting inflammation and fibrosis, MR overactivation constitutes a pivotal determinant of CKD progression and its associated morbidity and mortality. In accord with this mechanism of action, blockade of the MR is currently being investigated as a novel treatment regimen to slow the progression of CKD. The recently reported FIDELIO-DKD (FInerenone in reducing kiDnEy faiLure and dIsease prOgression in Diabetic Kidney Disease) study demonstrated that patients with CKD and type 2 diabetes who were treated with finerenone (a novel nonsteroidal MRA) manifested a lower risk of a composite primary outcome event compared with patients in the placebo arm (defined as kidney failure, or a sustained decrease of ≥40% in the estimated glomerular filtration rate from baseline, or death from renal causes). In addition, patients in the finerenone group also manifested a lower risk of a key secondary outcome event (defined as death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for heart failure). Key Messages: Based on the success of the FIDELIO-DKD study, future studies should be implemented testing the hypothesis that a wide array of nondiabetic CKD is modulated by overactivation of the MR, and consequently may be amenable to treatment with novel nonsteroidal MRAs. Future studies are encouraged to elucidate the clinical implications of the interplay of nonsteroidal MRAs and the components of the renin-angiotensin cascade. The unique and recently reported interrelationship of fibroblast growth factor (FGF23) and aldosterone may also constitute a propitious subject for future investigation.
Collapse
Affiliation(s)
- Murray Epstein
- University of Miami School of Medicine, Miami, Florida, USA
| |
Collapse
|
24
|
Yang J, Hu S, Huang L, Zhou J, Xiang H, Yang H, Cheng H, Tang Y. Protective effect of inhibiting TRPM7 expression on hypoxia post-treatment H9C2 cardiomyocytes. Clin Hemorheol Microcirc 2021; 77:91-105. [PMID: 32924998 DOI: 10.3233/ch-200934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Transient receptor potential channel 7 (TRPM7) plays an important role in maintaining intracellular ion concentration and osmotic pressure. OBJECTIVE The purpose of this study was to investigate the role and mechanism of inhibiting the expression of TRPM7 in the treatment of distal myocardial ischemia. METHODS H9C2 cells were treated with hypoxia post-treatment and reperfusion, respectively, detect the expression of HIF-1α and TRPM7, the concentration of Ca2+ and the degree of apoptosis in the H9C2 cells. The relevant miRNAs targeting TRPM7 were searched, the TRPM7 interference vectors were constructed, and the interference of different interference vectors on TRPM7 in H9C2 cells was detected. RESULTS The results showed that hypoxia post-treatment treatment would lead to increased expression of miR-22-3p which directly targeting TRPM7, decreased expression of TRPM7, increased expression of HIF-α and increased intracellular Ca2+ concentration. While reperfusion can increase the expression of HIF-1α and TRPM7 in H9C2 cells and increase the degree of apoptosis. CONCLUSION Knockdown of TRPM7 can significantly reduce reperfusion injury in H9C2 cells, reduce the degree of apoptosis, and the TRPM7 interference vector can inhibit the expression of TRPM7 and have a certain protective effect on the reperfusion injury of H9C2 cells.
Collapse
Affiliation(s)
- Juesheng Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, China
| | - Shan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, China
| | - Li Huang
- Medical Department of Graduate School of Nanchang University, Nanchang Jiangxi, China
| | - Jianliang Zhou
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, China
| | - Haiyan Xiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, China
| | - Heng Yang
- Medical Department of Graduate School of Nanchang University, Nanchang Jiangxi, China
| | - Hongzhang Cheng
- Medical Department of Graduate School of Nanchang University, Nanchang Jiangxi, China
| | - Yanhua Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang Jiangxi, China
| |
Collapse
|
25
|
Ferreira NS, Tostes RC, Paradis P, Schiffrin EL. Aldosterone, Inflammation, Immune System, and Hypertension. Am J Hypertens 2021; 34:15-27. [PMID: 32820797 PMCID: PMC7891246 DOI: 10.1093/ajh/hpaa137] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/15/2020] [Accepted: 08/17/2020] [Indexed: 12/23/2022] Open
Abstract
Aldosterone is a mineralocorticoid hormone that controls body fluid and electrolyte balance. Excess aldosterone is associated with cardiovascular and metabolic diseases. Inflammation plays a critical role on vascular damage promoted by aldosterone and aggravates vascular abnormalities, including endothelial dysfunction, vascular remodeling, fibrosis and oxidative stress, and other manifestations of end-organ damage that are associated with hypertension, other forms of cardiovascular disease, and diabetes mellitus and the metabolic syndrome. Over the past few years, many studies have consistently shown that aldosterone activates cells of the innate and adaptive immune systems. Macrophages and T cells accumulate in the kidneys, heart, and vasculature in response to aldosterone, and infiltration of immune cells contributes to end-organ damage in cardiovascular and metabolic diseases. Aldosterone activates various subsets of innate immune cells such as dendritic cells and monocytes/macrophages, as well as adaptive immune cells such as T lymphocytes, and, by activation of mineralocorticoid receptors stimulates proinflammatory transcription factors and the production of adhesion molecules and inflammatory cytokines and chemokines. This review will briefly highlight some of the studies on the involvement of aldosterone in activation of innate and adaptive immune cells and its impact on the cardiovascular system. Since aldosterone plays a key role in many cardiovascular and metabolic diseases, these data will open up promising perspectives for the identification of novel biomarkers and therapeutic targets for prevention and treatment of diseases associated with increased levels of aldosterone, such as arterial hypertension, obesity, the metabolic syndrome, and heart failure.
Collapse
Affiliation(s)
- Nathanne S Ferreira
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Pierre Paradis
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montréal, Québec, Canada
| |
Collapse
|
26
|
Zimmerman AD, Mackay L, Kemppainen RJ, Jones MA, Read CC, Schwartz D, Foradori CD. The Herbicide Atrazine Potentiates Angiotensin II-Induced Aldosterone Synthesis and Release From Adrenal Cells. Front Endocrinol (Lausanne) 2021; 12:697505. [PMID: 34335472 PMCID: PMC8317615 DOI: 10.3389/fendo.2021.697505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/22/2021] [Indexed: 12/06/2022] Open
Abstract
Atrazine is one of the most commonly used pre-emergence and early post-emergence herbicides in the world. We have shown previously that atrazine does not directly stimulate the pituitary or adrenal to trigger hormone release but acts centrally to activate a stress-like activation of the hypothalamic-pituitary-adrenal axis. In doing so, atrazine treatment has been shown to cause adrenal morphology changes characteristic of repeated stress. In this study, adrenals from atrazine treated and stressed animals were directly compared after 4 days of atrazine treatment or restraint stress. Both atrazine and stressed animals displayed reduced adrenocortical zona glomerulosa thickness and aldosterone synthase (CYP11B2) expression, indicative of repeated adrenal stimulation by adrenocorticotropic hormone. To determine if reduced CYP11B2 expression resulted in attenuated aldosterone synthesis, stressed and atrazine treated animals were challenged with angiotensin II (Ang II). As predicted, stressed animals produced less aldosterone compared to control animals when stimulated. However, atrazine treated animals had higher circulating aldosterone concentrations compared to both stressed and control groups. Ang II-induced aldosterone release was also potentiated in atrazine pretreated human adrenocortical carcinoma cells (H295R). Atrazine pretreated did not alter the expression of the rate limiting steroidogenic StAR protein or angiotensin II receptor 1. Atrazine treated animals also presented with higher basal blood pressure than vehicle treated control animals suggesting sustained elevations in circulating aldosterone levels. Our results demonstrate that treatment with the widely used herbicide, atrazine, directly increases stimulated production of aldosterone in adrenocortical cells independent of expression changes to rate limiting steroidogenic enzymes.
Collapse
|
27
|
Aujla PK, Kassiri Z. Diverse origins and activation of fibroblasts in cardiac fibrosis. Cell Signal 2020; 78:109869. [PMID: 33278559 DOI: 10.1016/j.cellsig.2020.109869] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
Cardiac fibroblasts (cFBs) have emerged as a heterogenous cell population. Fibroblasts are considered the main cell source for synthesis of the extracellular matrix (ECM) and as such a dysregulation in cFB function, activity, or viability can lead to disrupted ECM structure or fibrosis. Fibrosis can be initiated in response to different injuries and stimuli, and can be reparative (beneficial) or reactive (damaging). FBs need to be activated to myofibroblasts (MyoFBs) which have augmented capacity in synthesizing ECM proteins, causing fibrosis. In addition to the resident FBs in the myocardium, a number of other cells (pericytes, fibrocytes, mesenchymal, and hematopoietic cells) can transform into MyoFBs, further driving the fibrotic response. Multiple molecules including hormones, cytokines, and growth factors stimulate this process leading to generation of activated MyoFBs. Contribution of different cell types to cFBs and MyoFBs can result in an exponential increase in the number of MyoFBs and an accelerated pro-fibrotic response. Given the diversity of the cell sources, and the array of interconnected signalling pathways that lead to formation of MyoFBs and subsequently fibrosis, identifying a single target to limit the fibrotic response in the myocardium has been challenging. This review article will delineate the importance and relevance of fibroblast heterogeneity in mediating fibrosis in different models of heart failure and will highlight important signalling pathways implicated in myofibroblast activation.
Collapse
Affiliation(s)
- Preetinder K Aujla
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Cardiovascular Research Center, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
28
|
Corrêa JWDN, Prado CM, Riul ME, Araújo AV, Rossi MA, Bendhack LM. Reversion of cardiovascular remodelling in renovascular hypertensive 2K-1C rats by renin-angiotensin system inhibitors. Clin Exp Pharmacol Physiol 2020; 47:1965-1977. [PMID: 32688435 DOI: 10.1111/1440-1681.13384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Evaluate whether the RAS dual blockade would induce additional beneficial effects on cardiovascular remodelling when compared to monotherapy in renal hypertensive two kidneys-one clip (2K-1C) rats. METHODS Hypertensive 2K-1C and normotensive (2K) rats were treated for 14 days with submaximal doses of losartan (LOS), enalapril (ENA), losartan plus enalapril (LOS + ENA) or vehicle (water). Blood pressure and some parameters of cardiovascular remodelling were evaluated. RESULTS Systolic blood pressure (SBP) was higher in 2K-1C (209 ± 3 mm Hg, P < .05) than in 2K (113 ± 1 mm Hg) rats. There was an additional effect in 2K-1C treated with LOS + ENA (153 ± 9 mm Hg) on lowering SBP when compared to LOS (184 ± 12 mm Hg) or ENA (177 ± 9 mm Hg). None of the treatments had effect on SBP in 2K rats. In 2K-1C, cardiomyocyte hypertrophy was reduced by all treatments, although the cardiac hypertrophy indexes remained unchanged. 2K-1C aortas presented medial thickening that was partially reduced by the treatments. Intimal hyperplasia observed in 2K-1C (15.56 ± 0.89 µm vs 8.24 ± 0.80 µm) was reversed by ENA (9.52 ± 0.45 µm) or LOS + ENA (8.17 ± 0.53 µm). Collagen deposition was increased in 2K-1C hearts (1.77 ± 0.16 vs 1.28 ± 0.09) and aortas (8.1 ± 0.6 vs 5.2 ± 0.2). Treatment with LOS reduced (1.12 ± 0.14%) and ENA (0.81 ± 0.11%) or LOS + ENA (0.86 ± 0.11%) additionally diminished collagen only in 2K-1C hearts. CONCLUSIONS Submaximal doses of ACEi and/or ARB have inhibitory actions on cardiac remodelling and vascular hypertrophy not entirely dependent on their effects on blood pressure normalization in renovascular hypertensive rats. Combined therapy produced additional reduction in blood pressure than monotherapy despite a similar inhibition on cardiovascular remodelling.
Collapse
Affiliation(s)
| | - Cibele Maria Prado
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Maria Elena Riul
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Alice Valença Araújo
- Nucleus of Public Health, Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Vitória de Santo Antão, Brazil
| | - Marcos Antonio Rossi
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Lusiane Maria Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| |
Collapse
|
29
|
Left ventricular remodeling and dysfunction in primary aldosteronism. J Hum Hypertens 2020; 35:131-147. [PMID: 33067554 DOI: 10.1038/s41371-020-00426-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022]
Abstract
Primary aldosteronism (PA) is a common cause of secondary hypertension and is associated with worse cardiovascular outcomes. The elevated aldosterone in PA leads to left ventricular (LV) remodeling and dysfunction. In recent decades, clinical studies have demonstrated worse LV remodeling including increased LV mass and cardiac fibrosis in patients with PA compared to patients with essential hypertension. Several mechanisms may explain the process of aldosterone-induced LV remodeling, including directly profibrotic and hypertrophic effects of aldosterone on myocardium, increased reactive oxygen species and profibrotic molecules, dysregulation of extracellular matrix metabolism, endothelium dysfunction and circulatory macrophages activation. LV remodeling causes LV diastolic and systolic dysfunction, which may consequently lead to clinical complications such as heart failure, atrial fibrillation, ischemic heart disease, and other vascular events. Adequate treatment with adrenalectomy or medical therapy can improve LV remodeling and dysfunction in PA patients. In this review, we discuss the mechanisms of aldosterone-induced LV remodeling and provide an up-to-date review of clinical research about LV remodeling-related heart structural changes, cardiac dysfunction, and their clinical impacts on patients with PA.
Collapse
|
30
|
Liu Y, Lin Y, Zhang MM, Li XH, Liu YY, Zhao J, Shi L. The relationship of plasma renin, angiotensin, and aldosterone levels to blood pressure variability and target organ damage in children with essential hypertension. BMC Cardiovasc Disord 2020; 20:296. [PMID: 32546130 PMCID: PMC7298948 DOI: 10.1186/s12872-020-01579-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND To investigate the relationships of plasma renin, angiotensin, and aldosterone levels to blood pressure variability and target organ damage in children with essential hypertension. METHODS A case-control study was conducted on 132 children diagnosed with essential hypertension (103 males and 29 females with the mean age of 11.8 ± 2.4 years). The plasma RAAS levels were measured using the enhanced chemiluminescence method, the ambulatory blood pressure was monitored for 24 h, and then the average real variability (ARV) was calculated. Data on indicators were used for assessing cardiac and renal damages. The correlations of plasma renin, angiotensin, and aldosterone (RAAS) levels to blood pressure variability (BPV) and target organ damage (TOD) were studied. A comparison between the groups was conducted using SPSS 20. RESULTS Among the 132 children, 55 cases had target organ damage. The 24-h ARV and the daytime ARV of the systolic blood pressure of the high angiotensin II (AT II) group was significantly higher than that of the normal AT II group (t = 2.175, P = 0.031; t = 2.672, P = 0.009). Plasma AT II and aldosterone levels were significantly associated with the left ventricular mass index (r = 0.329, P = 0.0001; r = 0.175, P = 0.045). Linear regression analysis showed that AT II [β ± s.e. = 0.025 ± 0.006, 95% CI (0.013-0.038), P = 0.0001] and aldosterone [β ± s.e. = 0.021 ± 0.007, 95% CI (0.008-0.034), P = 0.002] were risk factors for LVH. CONCLUSIONS The AT II level in children with essential hypertension affected the variability of the 24-h and the daytime SBP. Plasma AT II and aldosterone levels were associated with cardiac damage. Results from this study indicated that AT II and aldosterone are risk factors for LVH in childhood hypertension and are of great significance for improving the clinical prognosis of pediatric patients with hypertension.
Collapse
Affiliation(s)
- Yang Liu
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100191, China
| | - Yao Lin
- Department of Cardiology, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ming-Ming Zhang
- Department of Cardiology, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing, 100020, China
| | - Xiao-Hui Li
- Department of Cardiology, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yan-Yan Liu
- Department of Cardiology, Children's Hospital Affiliated to Capital Institute of Pediatrics, Beijing, 100020, China
| | - Jing Zhao
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100191, China.
| | - Lin Shi
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100191, China.
| |
Collapse
|
31
|
Abstract
Primary aldosteronism (PA), the most common form of secondary hypertension, has been considered for decades as a "benign" form of hypertension, but evidences progressively built up to show that patients with PA had an excess rate of cardiovascular damage as compared to blood pressure-matched essential hypertensive patients. This review provides an updated view of structural and electrical cardiac remodeling and of vascular changes in hyperaldosteronism, and how they can favor development of cardiovascular events. The link between hyperaldosteronism and resistant hypertension is also examined, and the impact of targeted treatment of hyperaldosteronism on cardiovascular changes is finally discussed.
Collapse
|
32
|
Chang YY, Liao CW, Tsai CH, Chen CW, Pan CT, Chen ZW, Chen YL, Lin LC, Chang YR, Wu VC, Wu KD, Hung CS, Lin YH. Left Ventricular Dysfunction in Patients With Primary Aldosteronism: A Propensity Score-Matching Follow-Up Study With Tissue Doppler Imaging. J Am Heart Assoc 2019; 8:e013263. [PMID: 31718437 PMCID: PMC6915298 DOI: 10.1161/jaha.119.013263] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Background Primary aldosteronism is the most common cause of secondary hypertension and is associated with left ventricular hypertrophy. However, whether aldosterone excess is responsible for left ventricular (LV) diastolic dysfunction is unknown. Methods and Results We prospectively enrolled 129 patients with aldosterone‐producing adenoma and 120 patients with essential hypertension, and analyzed their clinical, biochemical, and echocardiographic data, including tissue Doppler images. The patients with aldosterone‐producing adenoma were reevaluated 1 year after adrenalectomy. After propensity score matching, there were 105 patients in each group. The patients with aldosterone‐producing adenoma had worse diastolic function than the patients with essential hypertension, as reflected by lower e′ (P<0.001) and higher E/e′ (P=0.003). Multivariate analysis showed that LV diastolic function was significantly correlated with age (P<0.001), sex (P<0.001), body mass index (P=0.002), systolic blood pressure (P=0.004), creatinine (P=0.008), and log‐transformed aldosterone‐renin ratio (P=0.003). After adrenalectomy, the patients with aldosterone‐producing adenoma had significant improvements in LV diastolic function as reflected by an increase in e′ (P=0.003) and decrease in E/e′ (P=0.002). The change in E/e′ was independently correlated with baseline E/e′ (P<0.001) and change in LV mass index (P=0.006). Conclusions The patients with primary aldosteronism had worse LV diastolic function than the patients with essential hypertension after propensity score matching, and this could be reversed after adrenalectomy, suggesting that aldosterone excess may induce LV diastolic dysfunction.
Collapse
Affiliation(s)
- Yi-Yao Chang
- Graduate Institute of Clinical Medicine National Taiwan University College of Medicine Taipei Taiwan.,Cardiology Division of Cardiovascular Medical Center Far Eastern Memorial Hospital New Taipei City Taiwan.,Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan.,Center of General Education Chihlee University of Technology New Taipei City Taiwan
| | - Che-Wei Liao
- Department of Internal Medicine National Taiwan University Hospital Hsin-Chu Branch Hsin-Chu Taiwan
| | - Cheng-Hsuan Tsai
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Ching-Way Chen
- Department of Internal Medicine National Taiwan University Hospital Yun-Lin Branch Yun-Lin Taiwan
| | - Chien-Ting Pan
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Zheng-Wei Chen
- Department of Internal Medicine National Taiwan University Hospital Yun-Lin Branch Yun-Lin Taiwan
| | - Ya-Li Chen
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Lung-Chun Lin
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Yi-Ru Chang
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Vin-Cent Wu
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Kwan-Dun Wu
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Chi-Sheng Hung
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | - Yen-Hung Lin
- Department of Internal Medicine National Taiwan University Hospital and National Taiwan University College of Medicine Taipei Taiwan
| | | | | |
Collapse
|
33
|
Dörr K, Kammer M, Reindl-Schwaighofer R, Lorenz M, Loewe C, Marculescu R, Erben R, Oberbauer R. Effect of etelcalcetide on cardiac hypertrophy in hemodialysis patients: a randomized controlled trial (ETECAR-HD). Trials 2019; 20:601. [PMID: 31651370 PMCID: PMC6813957 DOI: 10.1186/s13063-019-3707-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/06/2019] [Indexed: 02/15/2023] Open
Abstract
Background Fibroblast growth factor 23 (FGF23) is associated with left ventricular hypertrophy (LVH) in patients with chronic kidney disease, and calcimimetic therapy reduces plasma concentrations of FGF23. It remains unknown whether treatment with the calcimimetic etelcalcetide (ETL) reduces LVH in patients on hemodialysis. Methods/design This single-blinded randomized trial of 12 months duration will test the effects of ETL compared with alfacalcidol on LVH and cardiac fibrosis in maintenance hemodialysis patients with secondary hyperparathyroidism. Both treatment regimens will be titrated to equally suppress secondary hyperparathyroidism while alfacalcidol treatment causes an increase and ETL a decrease in FGF23, respectively. Patients treated thrice weekly with hemodialysis for ≥ 3 months and ≤ 3 years with parathyroid hormone levels ≥ 300 pg/ml and LVH will be enrolled in the study. The primary study endpoint is change from baseline to 12 months in left ventricular mass index (LVMI; g/m2) measured by cardiac magnetic resonance imaging. Sample size calculations showed that 62 randomized patients will be necessary to detect a difference in LVMI of at least 20 g/m2 between the two groups at 12 months. Due to the strong association of volume overload and LVH, randomization will be stratified by residual kidney function, and regular body composition monitoring will be performed to control the volume status of patients. Study medication will be administered intravenously by the dialysis nurses after every hemodialysis session, thus omitting adherence issues. Secondary study endpoints are cardiac parameters measured by echocardiography, biomarker concentrations of bone metabolism (FGF23, vitamin D, parathyroid hormone, calcium, phosphate, s-Klotho), cardiac markers (pro-brain natriuretic peptide, pre- and postdialysis troponin T) and metabolites of the renin–angiotensin–aldosterone cascade (angiotensin I (Ang I), Ang II, Ang-(1–7), Ang-(1–5), Ang-(1–9), and aldosterone). Discussion The causal inference and pathophysiology of LVH regression by FGF23 reduction using calcimimetic treatment has not yet been shown. This intervention study has the potential to discover a new strategy for the treatment of cardiac hypertrophy and fibrosis in patients on maintenance hemodialysis. It might be speculated that successful treatment of cardiac morphology will also reduce the risk of cardiac death in this population. Trial registration European Clinical Trials Database, EudraCT number 2017-000222-35; ClinicalTrials.gov, NCT03182699. Registered on
Collapse
Affiliation(s)
- Katharina Dörr
- Department of Nephrology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Michael Kammer
- Department of Nephrology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.,Center for Medical Statistics, Informatics and Intelligent Systems (CeMSIIS), Section for Clinical Biometrics, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | | | - Matthias Lorenz
- Vienna Dialysis Center, Kapellenweg 37, 1220, Vienna, Austria
| | - Christian Loewe
- Division of Cardiovascular and Interventional Radiology, Department of Bioimaging and Image-Guided Intervention, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Rodrig Marculescu
- Laboratory Medicine, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria
| | - Reinhold Erben
- Physiology, Pathophysiology, and Experimental Endocrinology, VetMed Vienna, Veterinärplatz 1, Vienna, Austria
| | - Rainer Oberbauer
- Department of Nephrology, Medical University of Vienna, Spitalgasse 23, 1090, Vienna, Austria.
| |
Collapse
|
34
|
Feng J, Armillei MK, Yu AS, Liang BT, Runnels LW, Yue L. Ca 2+ Signaling in Cardiac Fibroblasts and Fibrosis-Associated Heart Diseases. J Cardiovasc Dev Dis 2019; 6:E34. [PMID: 31547577 PMCID: PMC6956282 DOI: 10.3390/jcdd6040034] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/16/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiac fibrosis is the excessive deposition of extracellular matrix proteins by cardiac fibroblasts and myofibroblasts, and is a hallmark feature of most heart diseases, including arrhythmia, hypertrophy, and heart failure. This maladaptive process occurs in response to a variety of stimuli, including myocardial injury, inflammation, and mechanical overload. There are multiple signaling pathways and various cell types that influence the fibrogenesis cascade. Fibroblasts and myofibroblasts are central effectors. Although it is clear that Ca2+ signaling plays a vital role in this pathological process, what contributes to Ca2+ signaling in fibroblasts and myofibroblasts is still not wholly understood, chiefly because of the large and diverse number of receptors, transporters, and ion channels that influence intracellular Ca2+ signaling. Intracellular Ca2+ signals are generated by Ca2+ release from intracellular Ca2+ stores and by Ca2+ entry through a multitude of Ca2+-permeable ion channels in the plasma membrane. Over the past decade, the transient receptor potential (TRP) channels have emerged as one of the most important families of ion channels mediating Ca2+ signaling in cardiac fibroblasts. TRP channels are a superfamily of non-voltage-gated, Ca2+-permeable non-selective cation channels. Their ability to respond to various stimulating cues makes TRP channels effective sensors of the many different pathophysiological events that stimulate cardiac fibrogenesis. This review focuses on the mechanisms of Ca2+ signaling in fibroblast differentiation and fibrosis-associated heart diseases and will highlight recent advances in the understanding of the roles that TRP and other Ca2+-permeable channels play in cardiac fibrosis.
Collapse
Affiliation(s)
- Jianlin Feng
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Maria K Armillei
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Albert S Yu
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Bruce T Liang
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Loren W Runnels
- Department of Pharmacology, Rutgers, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| | - Lixia Yue
- Calhoun Cardiology Center, Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
35
|
Abstract
Primary aldosteronism (PA) is the most common form of secondary hypertension. In many cases, somatic mutations in ion channels and pumps within adrenal cells initiate the pathogenesis of PA, and this mechanism might explain why PA is so common and suggests that milder and evolving forms of PA must exist. Compared with primary hypertension, PA causes more end-organ damage and is associated with excess cardiovascular morbidity, including heart failure, stroke, nonfatal myocardial infarction, and atrial fibrillation. Screening is simple and readily available, and targeted therapy improves blood pressure control and mitigates cardiovascular morbidity. Despite these imperatives, screening rates for PA are low, and mineralocorticoid-receptor antagonists are underused for hypertension treatment. After the evidence for the prevalence of PA and its associated cardiovascular morbidity is summarized, a practical approach to PA screening, referral, and management is described. All physicians who treat hypertension should routinely screen appropriate patients for PA.
Collapse
Affiliation(s)
| | - Adina F Turcu
- Division of Metabolism, Endocrinology, and Diabetes (A.F.T., R.J.A.)
| | - Richard J Auchus
- Division of Metabolism, Endocrinology, and Diabetes (A.F.T., R.J.A.).,Department of Pharmacology (R.J.A.), University of Michigan, Ann Arbor
| |
Collapse
|
36
|
Cardiac fibrosis: potential therapeutic targets. Transl Res 2019; 209:121-137. [PMID: 30930180 PMCID: PMC6545256 DOI: 10.1016/j.trsl.2019.03.001] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 01/14/2023]
Abstract
Cardiovascular disease is a leading cause of mortality in the world and is exacerbated by the presence of cardiac fibrosis, defined by the accumulation of noncontractile extracellular matrix proteins. Cardiac fibrosis is directly linked to cardiac dysfunction and increased risk of arrhythmia. Despite its prevalence, there is a lack of efficacious therapies for inhibiting or reversing cardiac fibrosis, largely due to the complexity of the cell types and signaling pathways involved. Ongoing research has aimed to understand the mechanisms of cardiac fibrosis and develop new therapies for treating scar formation. Major approaches include preventing the formation of scar tissue and replacing fibrous tissue with functional cardiomyocytes. While targeting the renin-angiotensin-aldosterone system is currently used as the standard line of therapy for heart failure, there has been increased interest in inhibiting the transforming growth factor-β signaling pathway due its established role in cardiac fibrosis. Significant advances in cell transplantation therapy and biomaterials engineering have also demonstrated potential in regenerating the myocardium. Novel techniques, such as cellular direct reprogramming, and molecular targets, such as noncoding RNAs and epigenetic modifiers, are uncovering novel therapeutic options targeting fibrosis. This review provides an overview of current approaches and discuss future directions for treating cardiac fibrosis.
Collapse
|
37
|
Ames MK, Atkins CE, Pitt B. The renin-angiotensin-aldosterone system and its suppression. J Vet Intern Med 2019; 33:363-382. [PMID: 30806496 PMCID: PMC6430926 DOI: 10.1111/jvim.15454] [Citation(s) in RCA: 268] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
Chronic activation of the renin-angiotensin-aldosterone system (RAAS) promotes and perpetuates the syndromes of congestive heart failure, systemic hypertension, and chronic kidney disease. Excessive circulating and tissue angiotensin II (AngII) and aldosterone levels lead to a pro-fibrotic, -inflammatory, and -hypertrophic milieu that causes remodeling and dysfunction in cardiovascular and renal tissues. Understanding of the role of the RAAS in this abnormal pathologic remodeling has grown over the past few decades and numerous medical therapies aimed at suppressing the RAAS have been developed. Despite this, morbidity from these diseases remains high. Continued investigation into the complexities of the RAAS should help clinicians modulate (suppress or enhance) components of this system and improve quality of life and survival. This review focuses on updates in our understanding of the RAAS and the pathophysiology of AngII and aldosterone excess, reviewing what is known about its suppression in cardiovascular and renal diseases, especially in the cat and dog.
Collapse
Affiliation(s)
- Marisa K Ames
- Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, Fort Collins, Colorado
| | - Clarke E Atkins
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Bertram Pitt
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan
| |
Collapse
|
38
|
Bartlett RS, Gaston JD, Ye S, Kendziorski C, Thibeault SL. Mechanotransduction of vocal fold fibroblasts and mesenchymal stromal cells in the context of the vocal fold mechanome. J Biomech 2018; 83:227-234. [PMID: 30553439 DOI: 10.1016/j.jbiomech.2018.11.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/03/2018] [Accepted: 11/30/2018] [Indexed: 01/20/2023]
Abstract
The design of cell-based therapies for vocal fold tissue engineering requires an understanding of how cells adapt to the dynamic mechanical forces found in the larynx. Our objective was to compare mechanotransductive processes in therapeutic cell candidates (mesenchymal stromal cells from adipose tissue and bone marrow, AT-MSC and BM-MSC) to native cells (vocal fold fibroblasts-VFF) in the context of vibratory strain. A bioreactor was used to expose VFF, AT-MSC, and BM-MSC to axial tensile strain and vibration at human physiological levels. Microarray, an empirical Bayes statistical approach, and geneset enrichment analysis were used to identify significant mechanotransductive pathways associated with the three cell types and three mechanical conditions. Two databases (Gene Ontology, Kyoto Encyclopedia of Genes and Genomes) were used for enrichment analyses. VFF shared more mechanotransductive pathways with BM-MSC than with AT-MSC. Gene expression that appeared to distinguish the vibratory strain condition from polystyrene condition for these two cells types related to integrin activation, focal adhesions, and lamellipodia activity, suggesting that vibratory strain may be associated with cytoarchitectural rearrangement, cell reorientation, and extracellular matrix remodeling. In response to vibration and tensile stress, BM-MSC better mimicked VFF mechanotransduction than AT-MSC, providing support for the consideration of BM-MSC as a cell therapy for vocal fold tissue engineering. Future research is needed to better understand the sorts of physical adaptations that are afforded to vocal fold tissue as a result of focal adhesions, integrins, and lamellipodia, and how these adaptations could be exploited for tissue engineering.
Collapse
Affiliation(s)
- Rebecca S Bartlett
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Wisconsin-Madison, Madison, WI, United States
| | - Joel D Gaston
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Wisconsin-Madison, Madison, WI, United States
| | - Shuyun Ye
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States
| | - Christina Kendziorski
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States
| | - Susan L Thibeault
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
39
|
Cardiac hypertrophy in chronic kidney disease—role of Aldosterone and FGF23. RENAL REPLACEMENT THERAPY 2018. [DOI: 10.1186/s41100-018-0152-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
40
|
Syed M, Ball JP, Mathis KW, Hall ME, Ryan MJ, Rothenberg ME, Yanes Cardozo LL, Romero DG. MicroRNA-21 ablation exacerbates aldosterone-mediated cardiac injury, remodeling, and dysfunction. Am J Physiol Endocrinol Metab 2018; 315:E1154-E1167. [PMID: 30153065 PMCID: PMC6336952 DOI: 10.1152/ajpendo.00155.2018] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 07/30/2018] [Accepted: 08/24/2018] [Indexed: 12/21/2022]
Abstract
Primary aldosteronism is characterized by excess aldosterone secretion by the adrenal gland independent of the renin-angiotensin system and accounts for ~10% of hypertensive patients. Excess aldosterone causes cardiac hypertrophy, fibrosis, inflammation, and hypertension. The molecular mechanisms that trigger the onset and progression of aldosterone-mediated cardiac injury remain incompletely understood. MicroRNAs (miRNAs) are endogenous, small, noncoding RNAs that have been implicated in multiple cardiac pathologies; however, their regulation and role in aldosterone-mediated cardiac injury and dysfunction remains mostly unknown. We previously reported that microRNA-21 (miR-21) is the most upregulated miRNA by excess aldosterone in the left ventricle in a rat experimental model of primary aldosteronism. To elucidate the role of miR-21 in aldosterone-mediated cardiac injury and dysfunction, miR-21 knockout mice and their wild-type littermates were treated with aldosterone infusion and salt in the drinking water for 2 or 8 wk. miR-21 genetic ablation exacerbated aldosterone/salt-mediated cardiac hypertrophy and cardiomyocyte cross-sectional area. Furthermore, miR-21 genetic ablation increased the cardiac expression of fibrosis and inflammation markers and fetal gene program. miR-21 genetic ablation increased aldosterone/salt-mediated cardiac dysfunction but did not affect aldosterone/salt-mediated hypertension. miR-21 target gene Sprouty 2 may be implicated in the cardiac effects of miR-21 genetic ablation. Our study shows that miR-21 genetic ablation exacerbates aldosterone/salt-mediated cardiac hypertrophy, injury, and dysfunction blood pressure independently. These results suggest that miR-21 plays a protective role in the cardiac pathology triggered by excess aldosterone. Furthermore, miR-21 supplementation may be a novel therapeutic approach to abolish or mitigate excess aldosterone-mediated cardiovascular deleterious effects in primary aldosteronism.
Collapse
Affiliation(s)
- Maryam Syed
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jana P Ball
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Keisa W Mathis
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Michael E Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Department of Medicine, University of Mississippi Medical Center , Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- G.V. (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine , Cincinnati, Ohio
| | - Licy L Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Medicine, University of Mississippi Medical Center , Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center , Jackson, Mississippi
| | - Damian G Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
- Women's Health Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Cardio Renal Research Center, University of Mississippi Medical Center , Jackson, Mississippi
- Mississippi Center for Excellence in Perinatal Research, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
41
|
Rouet‐Benzineb P, Merval R, Polidano E. Effects of hypoestrogenism and/or hyperaldosteronism on myocardial remodeling in female mice. Physiol Rep 2018; 6:e13912. [PMID: 30430766 PMCID: PMC6236131 DOI: 10.14814/phy2.13912] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/12/2018] [Accepted: 10/05/2018] [Indexed: 12/18/2022] Open
Abstract
We investigated the potential adverse effects of hyperaldosteronism and/or hypoestrogenism on cardiac phenotype, and examined their combined effects in female mice overexpressing cardiac aldosterone synthase (AS). We focused on some signaling cascades challenging defensive responses to adapt and/or to survive in the face of double deleterious stresses, such as Ca2+ -homeostasis, pro/anti-hypertrophic, endoplasmic reticulum stress (ER stress), pro- or anti-apoptotic effectors, and MAP kinase activation, and redox signaling. These protein expressions were assessed by immunoblotting at 9 weeks after surgery. Female wild type (FWT) and FAS mice were fed with phytoestrogen-free diet; underwent ovariectomy (Ovx) or sham-operation (Sham). Ovx increased gain weight and hypertrophy index. Transthoracic echocardiograghy was performed. Both Ovx-induced heart rate decrease and fractional shortening increase were associated with collagen type III shift. Cardiac estrogen receptor (ERα, ERβ) protein expression levels were downregulated in Ovx mice. Hypoestrogenism increased plasma aldosterone and MR protein expression in FAS mice. Both aldosterone and Ovx played as mirror effects on up and downstream signaling effectors of calcium/redox homeostasis, apoptosis, such as concomitant CaMKII activation and calcineurin down-regulation, MAP kinase inhibition (ERK1/2, p38 MAPK) and Akt activation. The ratio Bcl2/Bax is in favor to promote cell survivor. Finally, myocardium had dynamically orchestrated multiple signaling cascades to restore tolerance to hostile environment thereby contributing to a better maintenance of Ca2+ /redox homeostasis. Ovx-induced collagen type III isoform shift and its upregulation may be important for the biomechanical transduction of the heart and the recovery of cardiac function in FAS mice. OVX antagonized aldosterone signaling pathways.
Collapse
|
42
|
Frangogiannis NG. Fibroblasts and the extracellular matrix in right ventricular disease. Cardiovasc Res 2018; 113:1453-1464. [PMID: 28957531 DOI: 10.1093/cvr/cvx146] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/01/2017] [Indexed: 12/17/2022] Open
Abstract
Right ventricular failure predicts adverse outcome in patients with pulmonary hypertension (PH), and in subjects with left ventricular heart failure and is associated with interstitial fibrosis. This review manuscript discusses the cellular effectors and molecular mechanisms implicated in right ventricular fibrosis. The right ventricular interstitium contains vascular cells, fibroblasts, and immune cells, enmeshed in a collagen-based matrix. Right ventricular pressure overload in PH is associated with the expansion of the fibroblast population, myofibroblast activation, and secretion of extracellular matrix proteins. Mechanosensitive transduction of adrenergic signalling and stimulation of the renin-angiotensin-aldosterone cascade trigger the activation of right ventricular fibroblasts. Inflammatory cytokines and chemokines may contribute to expansion and activation of macrophages that may serve as a source of fibrogenic growth factors, such as transforming growth factor (TGF)-β. Endothelin-1, TGF-βs, and matricellular proteins co-operate to activate cardiac myofibroblasts, and promote synthesis of matrix proteins. In comparison with the left ventricle, the RV tolerates well volume overload and ischemia; whether the right ventricular interstitial cells and matrix are implicated in these favourable responses remains unknown. Expansion of fibroblasts and extracellular matrix protein deposition are prominent features of arrhythmogenic right ventricular cardiomyopathies and may be implicated in the pathogenesis of arrhythmic events. Prevailing conceptual paradigms on right ventricular remodelling are based on extrapolation of findings in models of left ventricular injury. Considering the unique embryologic, morphological, and physiologic properties of the RV and the clinical significance of right ventricular failure, there is a need further to dissect RV-specific mechanisms of fibrosis and interstitial remodelling.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B Bronx, 10461 NY, USA
| |
Collapse
|
43
|
Liu Q, Song J, Lu D, Geng J, Jiang Z, Wang K, Zhang B, Shan Q. Effects of renal denervation on monocrotaline induced pulmonary remodeling. Oncotarget 2018; 8:46846-46855. [PMID: 28187460 PMCID: PMC5564527 DOI: 10.18632/oncotarget.15154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/24/2017] [Indexed: 12/18/2022] Open
Abstract
Pulmonary artery hypertension (PAH) is a rapidly progressive disorder, which leads to right heart failure and even death. Overactivity of the renin-angiotensin-aldosterone system (RAAS) and sympathetic nervous system accounts for the development and progression of PAH. The role of renal denervation (RDN) in different periods of PAH has not been fully elucidated. A single intraperitoneal injection of monocrotaline (MCT, 60 mg/kg) was used to induce pulmonary remodeling in male Sprague Dawley rats (n = 40). After 24-hour of MCT administration, a subset of rats underwent RDN (RDN24h, n = 10); after 2-week of MCT injection, another ten rats received RDN treatment (RDN2w, n = 10) and the left 20 rats were divided to MCT group with sham RDN operation (MCT, n = 20). Eight rats in Control group received intraperitoneal injection of normal saline (60 mg/kg) once and sham RDN surgery. After 35 days, tissue and blood samples were collected. Histological analysis demonstrated that the collagen volume fraction of right ventricle, lung tissue and pulmonary vessel reduced significantly in RDN24h group but not in the RDN2w group, compared with MCT group. Moreover, the earlier RDN treatment significantly decreased SNS activity and blunted RAAS activation. Importantly, RDN treatment significantly improved the survival rate. In summary, earlier RDN treatment could attenuate cardio-pulmonary fibrosis and therefore might be a promising approach to prevent the development of PAH.
Collapse
Affiliation(s)
- Qian Liu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiyang Song
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, China
| | - Dasheng Lu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Geng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixin Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Kai Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bin Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qijun Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
44
|
Hundeshagen G, Herndon DN, Clayton RP, Wurzer P, McQuitty A, Jennings K, Branski L, Collins VN, Marques NR, Finnerty CC, Suman OE, Kinsky MP. Long-term effect of critical illness after severe paediatric burn injury on cardiac function in adolescent survivors: an observational study. THE LANCET. CHILD & ADOLESCENT HEALTH 2017; 1:293-301. [PMID: 29581998 PMCID: PMC5865217 DOI: 10.1016/s2352-4642(17)30122-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Sepsis, trauma, and burn injury acutely depress systolic and diastolic cardiac function; data on long-term cardiac sequelae of pediatric critical illness are sparse. This study evaluated long-term systolic and diastolic function, myocardial fibrosis, and exercise tolerance in survivors of severe pediatric burn injury. METHODS Subjects at least 5 years after severe burn (post-burn:PB) and age-matched healthy controls (HC) underwent echocardiography to quantify systolic function (ejection fraction[EF%]), diastolic function (E/e'), and myocardial fibrosis (calibrated integrated backscatter) of the left ventricle. Exercise tolerance was quantified by oxygen consumption (VO2) and heart rate at rest and peak exercise. Demographic information, clinical data, and biomarker expression were used to predict long-term cardiac dysfunction and fibrosis. FINDINGS Sixty-five subjects (PB:40;HC:25) were evaluated. At study date, PB subjects were 19±5 years, were at 12±4 years postburn, and had burns over 59±19% of total body surface area, sustained at 8±5 years of age. The PB group had lower EF% (PB:52±9%;HC:61±6%; p=0.004), E/e' (PB:9.8±2.9;HC: 5.4±0.9;p<0.0001), VO2peak (PB:37.9±12;HC: 46±8.32 ml/min/kg; p=0.029), and peak heart rate (PB:161±26;HC:182±13bpm;p=0.007). The PB group had moderate (28%) or severe (15%) systolic dysfunction, moderate (50%) or severe diastolic dysfunction (21%), and myocardial fibrosis (18%). Biomarkers and clinical parameters predicted myocardial fibrosis, systolic dysfunction, and diastolic dysfunction. INTERPRETATION Severe pediatric burn injury may have lasting impact on cardiac function into young adulthood and is associated with myocardial fibrosis and reduced exercise tolerance. Given the strong predictive value of systolic and diastolic dysfunction, these patients might be at increased risk for early heart failure, associated morbidity, and mortality. FUNDING Conflicts of Interest and Sources of Funding: The authors do not have any conflicts of interest to declare. This work was supported by NIH (P50 GM060338, R01 GM056687, R01 HD049471, R01 GM112936, R01-GM56687 and T32 GM008256), NIDILRR (H133A120091, 90DP00430100), Shriners Hospitals for Children (84080, 79141, 79135, 71009, 80100, 71008, 87300 and 71000), FAER (MRTG CON14876), and the Department of Defense (W81XWH-14-2-0162 and W81XWH1420162). It was also made possible with the support of UTMB's Institute for Translational Sciences, supported in part by a Clinical and Translational Science Award (UL1TR000071) from the National Center for Advancing Translational Sciences (NIH).
Collapse
Affiliation(s)
- Gabriel Hundeshagen
- Department of Surgery, University of Texas Medical Branch, 301
University Blvd, Galveston, TX 77555
- Shriners Hospitals for Children, Galveston, 815 Market St,
Galveston, TX 77555
- Department of Hand, Plastic and Reconstructive Surgery, Burn Trauma
Center, BG Trauma Center Ludwigshafen; University of Heidelberg,
Ludwig-Guttmann-Str. 13, 67071 Ludwigshafen, Germany
| | - David N Herndon
- Department of Surgery, University of Texas Medical Branch, 301
University Blvd, Galveston, TX 77555
- Shriners Hospitals for Children, Galveston, 815 Market St,
Galveston, TX 77555
| | - Robert P Clayton
- Department of Surgery, University of Texas Medical Branch, 301
University Blvd, Galveston, TX 77555
- Shriners Hospitals for Children, Galveston, 815 Market St,
Galveston, TX 77555
| | - Paul Wurzer
- Division of Plastic, Aesthetic and Reconstructive Surgery,
Department of Surgery, Medical University of Graz, Austria
| | - Alexis McQuitty
- Department of Anesthesiology, University of Texas Medical Branch,
301 University Blvd, Galveston, TX 77555
| | - Kristofer Jennings
- Office of Biostatistics, Department of Preventive Medicine and
Community Health, University of Texas Medical Branch, 301 University Blvd,
Galveston, TX 77555
| | - Ludwik Branski
- Department of Surgery, University of Texas Medical Branch, 301
University Blvd, Galveston, TX 77555
- Shriners Hospitals for Children, Galveston, 815 Market St,
Galveston, TX 77555
- Division of Plastic, Aesthetic and Reconstructive Surgery,
Department of Surgery, Medical University of Graz, Austria
| | - Vanessa N Collins
- Shriners Hospitals for Children, Galveston, 815 Market St,
Galveston, TX 77555
| | - Nicole Ribeiro Marques
- Department of Anesthesiology, University of Texas Medical Branch,
301 University Blvd, Galveston, TX 77555
| | - Celeste C Finnerty
- Department of Surgery, University of Texas Medical Branch, 301
University Blvd, Galveston, TX 77555
- Shriners Hospitals for Children, Galveston, 815 Market St,
Galveston, TX 77555
| | - Oscar E Suman
- Department of Surgery, University of Texas Medical Branch, 301
University Blvd, Galveston, TX 77555
- Shriners Hospitals for Children, Galveston, 815 Market St,
Galveston, TX 77555
| | - Michael P Kinsky
- Division of Plastic, Aesthetic and Reconstructive Surgery,
Department of Surgery, Medical University of Graz, Austria
| |
Collapse
|
45
|
Ivey MJ, Kuwabara JT, Pai JT, Moore RE, Sun Z, Tallquist MD. Resident fibroblast expansion during cardiac growth and remodeling. J Mol Cell Cardiol 2017; 114:161-174. [PMID: 29158033 DOI: 10.1016/j.yjmcc.2017.11.012] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/25/2017] [Accepted: 11/16/2017] [Indexed: 01/18/2023]
Abstract
Cardiac fibrosis, denoted by the deposition of extracellular matrix, manifests with a variety of diseases such as hypertension, diabetes, and myocardial infarction. Underlying this pathological extracellular matrix secretion is an expansion of fibroblasts. The mouse is now a common experimental model system for the study of cardiovascular remodeling and elucidation of fibroblast responses to cardiac growth and stress is vital for understanding disease processes. Here, using diverse but fibroblast specific markers, we report murine fibroblast distribution and proliferation in early postnatal, adult, and injured hearts. We find that perinatal fibroblasts and endothelial cells proliferate at similar rates. Furthermore, regardless of the injury model, fibroblast proliferation peaks within the first week after injury, a time window similar to the period of the inflammatory phase. In addition, fibroblast densities remain high weeks after the initial insult. These results provide detailed information regarding fibroblast distribution and proliferation in experimental methods of heart injury.
Collapse
Affiliation(s)
- Malina J Ivey
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States
| | - Jill T Kuwabara
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States
| | - Jonathan T Pai
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States
| | - Richard E Moore
- Department of Molecular Biochemistry and Bioengineering, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States
| | - Zuyue Sun
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States
| | - Michelle D Tallquist
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, United States.
| |
Collapse
|
46
|
Belden Z, Deiuliis JA, Dobre M, Rajagopalan S. The Role of the Mineralocorticoid Receptor in Inflammation: Focus on Kidney and Vasculature. Am J Nephrol 2017; 46:298-314. [PMID: 29017166 PMCID: PMC6863172 DOI: 10.1159/000480652] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The remarkable success of clinical trials in mineralocorticoid receptor (MR) inhibition in heart failure has driven research on the physiological and pathological role(s) of nonepithelial MR expression. MR is widely expressed in the cardiovascular system and is a major determinant of endothelial function, smooth muscle tone, vascular remodeling, fibrosis, and blood pressure. An important new dimension is the appreciation of the role MR plays in immune cells and target organ damage in the heart, kidney and vasculature, and in the development of insulin resistance. SUMMARY The mechanism for MR activation in tissue injury continues to evolve with the evidence to date suggesting that activation of MR results in a complex repertoire of effects involving both macrophages and T cells. MR is an important transcriptional regulator of macrophage phenotype and function. Another important feature of MR activation is that it can occur even with normal or low aldosterone levels in pathological conditions. Tissue-specific conditional models of MR expression in myeloid cells, endothelial cells, smooth muscle cells and cardiomyocytes have been very informative and have firmly demonstrated a critical role of MR as a key pathophysiologic variable in cardiac hypertrophy, transition to heart failure, adipose inflammation, and atherosclerosis. Finally, the central nervous system activation of MR in permeable regions of the blood-brain barrier may play a role in peripheral inflammation. Key Message: Ongoing clinical trials will help clarify the role of MR blockade in conditions, such as atherosclerosis and chronic kidney disease.
Collapse
Affiliation(s)
- Zachary Belden
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jeffrey A. Deiuliis
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mirela Dobre
- Division of Nephrology and Hypertension, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sanjay Rajagopalan
- Case Cardiovascular Research Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Harrington Heart and Vascular Institute, Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
47
|
Kuwabara JT, Tallquist MD. Tracking Adventitial Fibroblast Contribution to Disease: A Review of Current Methods to Identify Resident Fibroblasts. Arterioscler Thromb Vasc Biol 2017; 37:1598-1607. [PMID: 28705796 DOI: 10.1161/atvbaha.117.308199] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 06/23/2017] [Indexed: 01/18/2023]
Abstract
Cells present in the adventitia, or outermost layer of the blood vessel, contribute to the progression of vascular diseases, such as atherosclerosis, hypertension, and aortic dissection. The adventitial fibroblast of the aorta is the prototypic perivascular fibroblast, but the adventitia is composed of multiple distinct cell populations. Therefore, methods for uniquely identifying the fibroblast are critical for a better understanding of how these cells contribute to disease processes. A popular method for distinguishing adventitial cell types relies on the use of genetic tools in the mouse to trace and manipulate these cells. Because lineage tracing relying on Cre-recombinase expressing mice is used more frequently in studies of vascular disease, it is important to outline the advantages and limitations of these genetic tools. The purpose of this article is to provide an overview of the various genetic tools available in the mouse for the study of resident adventitial fibroblasts.
Collapse
Affiliation(s)
- Jill T Kuwabara
- From the Center for Cardiovascular Research, University of Hawaii, Honolulu
| | | |
Collapse
|
48
|
Kolkhof P, Bärfacker L. 30 YEARS OF THE MINERALOCORTICOID RECEPTOR: Mineralocorticoid receptor antagonists: 60 years of research and development. J Endocrinol 2017; 234:T125-T140. [PMID: 28634268 PMCID: PMC5488394 DOI: 10.1530/joe-16-0600] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 02/21/2017] [Indexed: 12/21/2022]
Abstract
The cDNA of the mineralocorticoid receptor (MR) was cloned 30 years ago, in 1987. At that time, spirolactone, the first generation of synthetic steroid-based MR antagonists (MRAs), which was identified in preclinical in vivo models, had already been in clinical use for 30 years. Subsequent decades of research and development by Searle & Co., Ciba-Geigy, Roussel Uclaf and Schering AG toward identifying a second generation of much more specific steroidal MRAs were all based on the initial 17-spirolactone construct. The salient example is eplerenone, first described in 1987, coincidentally with the cloning of MR cDNA. Its launch on the market in 2003 paralleled intensive drug discovery programs for a new generation of non-steroidal MRAs. Now, 30 years after the cDNA cloning of MR and 60 years of clinical use of steroidal MRAs, novel non-steroidal MRAs such as apararenone, esaxerenone and finerenone are in late-stage clinical trials in patients with heart failure, chronic kidney disease (CKD), hypertension and liver disease. Finerenone has already been studied in over 2000 patients with heart failure plus chronic kidney disease and/or diabetes, and in patients with diabetic kidney disease, in five phase II clinical trials. Here, we reflect on the history of the various generations of MRAs and review characteristics of the most important steroidal and non-steroidal MRAs.
Collapse
Affiliation(s)
- Peter Kolkhof
- Drug DiscoveryCardiology Research, Bayer AG, Wuppertal, Germany
| | - Lars Bärfacker
- Drug DiscoveryMedicinal Chemistry, Bayer AG, Wuppertal, Germany
| |
Collapse
|
49
|
Ball JP, Syed M, Marañon RO, Hall ME, KC R, Reckelhoff JF, Yanes Cardozo LL, Romero DG. Role and Regulation of MicroRNAs in Aldosterone-Mediated Cardiac Injury and Dysfunction in Male Rats. Endocrinology 2017; 158:1859-1874. [PMID: 28368454 PMCID: PMC5460923 DOI: 10.1210/en.2016-1707] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 03/15/2017] [Indexed: 12/21/2022]
Abstract
Primary aldosteronism is characterized by excess aldosterone (ALDO) secretion independent of the renin-angiotensin system and accounts for approximately 10% of hypertension cases. Excess ALDO that is inappropriate for salt intake status causes cardiac hypertrophy, inflammation, fibrosis, and hypertension. The molecular mechanisms that trigger the onset and progression of ALDO-mediated cardiac injury are poorly understood. MicroRNAs (miRNAs) are endogenous, small, noncoding RNAs that have been implicated in diverse cardiac abnormalities, yet very little is known about their regulation and role in ALDO-mediated cardiac injury. To elucidate the regulation of miRNAs in ALDO-mediated cardiac injury, we performed a time-series analysis of left ventricle (LV) miRNA expression. Uninephrectomized male Sprague-Dawley rats were treated with ALDO (0.75 µg/h) infusion and SALT (1.0% NaCl/0.3% KCl) in the drinking water for up to 8 weeks. ALDO/SALT time dependently modulated the expression of multiple miRNAs in the LV. miR-21 was the most upregulated miRNA after 2 weeks of treatment and remained elevated until the end of the study. To elucidate the role of miR-21 in ALDO/SALT-mediated cardiac injury, miR-21 was downregulated by using antagomirs in ALDO/SALT-treated rats. miR-21 downregulation exacerbated ALDO/SALT-mediated cardiac hypertrophy, expression of fibrosis marker genes, interstitial and perivascular fibrosis, OH-proline content, and cardiac dysfunction. These results suggest that ALDO/SALT-mediated cardiac miR-21 upregulation may be a compensatory mechanism that mitigates ALDO/SALT-mediated cardiac deleterious effects. We speculate that miR-21 supplementation would have beneficial effects in reverting or mitigating cardiac injury and dysfunction in patients with primary aldosteronism.
Collapse
Affiliation(s)
- Jana P. Ball
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Maryam Syed
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Rodrigo O. Marañon
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Michael E. Hall
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Roshan KC
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Jane F. Reckelhoff
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Licy L. Yanes Cardozo
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Damian G. Romero
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Cardio-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216
| |
Collapse
|
50
|
BNIP3L promotes cardiac fibrosis in cardiac fibroblasts through [Ca 2+] i-TGF-β-Smad2/3 pathway. Sci Rep 2017; 7:1906. [PMID: 28507335 PMCID: PMC5432493 DOI: 10.1038/s41598-017-01936-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/03/2017] [Indexed: 01/17/2023] Open
Abstract
Fibrosis is an important, structurally damaging event that occurs in pathological cardiac remodeling, leading to cardiac dysfunction. BNIP3L is up-regulated in pressure overload-induced heart failure and has been reported to play an important role in cardiomyocyte apoptosis; however, its involvement in cardiac fibroblasts (CFs) remains unknown. We prove for the first time that the expression of BNIP3L is significantly increased in the CFs of rats undergoing pressure overload-induced heart failure. Furthermore, this increased BNIP3L expression was confirmed in cultured neonatal rat CFs undergoing proliferation and extracellular matrix (ECM) protein over-expression that was induced by norepinephrine (NE). The overexpression or suppression of BNIP3L promoted or inhibited NE-induced proliferation and ECM expression in CFs, respectively. In addition, [Ca2+]i, transforming growth factor beta (TGF-β) and the nuclear accumulation of Smad2/3 were successively increased when BNIP3L was overexpressed and reduced when BNIP3L was inhibited. Furthermore, the down-regulation of TGF-β by TGF-β-siRNA attenuated the increase of BNIP3L-induced fibronectin expression. We also demonstrated that the increase of BNIP3L in CFs was regulated by NE-AR-PKC pathway in vitro and in vivo. These results reveal that BNIP3L is a novel mediator of pressure overload-induced cardiac fibrosis through the [Ca2+]i-TGF-β-Smad2/3 pathway in CFs.
Collapse
|