1
|
Aleshcheva G, Salih S, Baumeier C, Escher F, Bock CT, Schultheiss HP. Discovery of miRNAs unique to actively transcribed erythroparvovirus infection in heart failure patients. ESC Heart Fail 2025. [PMID: 39970057 DOI: 10.1002/ehf2.15194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/26/2024] [Accepted: 12/09/2024] [Indexed: 02/21/2025] Open
Abstract
AIMS miRNAs, small non-coding RNAs, play key roles in gene regulation, cell differentiation and tissue development. They influence viral infection outcomes by directly interacting with viral genomes or modifying the host microenvironment. This study demonstrates miRNAs' ability to selectively suppress transcriptionally active erythroparvovirus, highlighting their potential in antiviral therapies. METHODS AND RESULTS Seventy-five endomyocardial biopsy (EMB) specimens from patients with unexplained heart failure were analysed. The samples included 19 with dilated cardiomyopathy and inflammation (DCMi), 12 with dilated cardiomyopathy (DCM), 25 with inflammation and active erythroparvovirus infection, 13 with active erythroparvovirus infection only and 6 from undiagnosed patients as controls. miRNA expression was measured using TaqMan assays. miR-98, miR-222, miR-106b and miR-197 were significantly upregulated in patients with transcriptionally active erythroparvovirus infection, independent of inflammation (P < 0.005). These miRNAs differentiated these patients from all other groups with over 90% specificity. CONCLUSIONS These specific miRNAs offer a novel diagnostic tool for active erythroparvovirus infections and hold promise as therapeutic targets, providing safer alternatives to traditional antiviral treatments.
Collapse
Affiliation(s)
- Ganna Aleshcheva
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany
| | - Sara Salih
- BHT - Berliner Hochschule für Technik, Berlin, Germany
| | | | - Felicitas Escher
- Institute for Cardiac Diagnostics and Therapy (IKDT), Berlin, Germany
- DHZC (German Heart Centre of Charité), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - C Thomas Bock
- Department of Infectious Diseases, Division of Viral Gastroenteritis and Hepatitis Pathogens and Enteroviruses, Robert Koch Institute, Berlin, Germany
| | | |
Collapse
|
2
|
Wang K, Li K, Li Z, Yan X. Circulating miRNA-21 as early potential diagnostic biomarker for acute myocardial infarction: a meta-analysis. Front Cardiovasc Med 2024; 11:1330884. [PMID: 39238499 PMCID: PMC11374624 DOI: 10.3389/fcvm.2024.1330884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
Introduction There exists a knowledge gap concerning the clinical significance of miRNA-21; therefore, in the present study, we aimed to estimate the diagnostic and prognostic accuracy and sensitivity of miRNA-21 in acute myocardial infarction (AMI) by performing an evidence-based meta-analysis of previous AMI-related clinical studies. Methods Chinese and English literature published before April 2024 were searched, and data were reviewed and extracted. After quality appraisal, the STATA 16.0 software was used for the effect size analysis of the various treatments described in the literature. Results A total of 14 valid documents were retrieved from 562 studies. The results of the systematic review revealed that for the patients with AMI vs. those without non-AMI, the aggregated odds ratio reached 5.37 (95% confidence interval 3.70-7.04). The general sensitivity and specificity for the circulating miRNA-21 levels in diagnosing AMI were 0.83 and 0.81, respectively. Discussion Thus, the meta-analysis of 14 AMI-related clinical trials highlighted that miRNA-21 may serve as a promising biomarker for diagnosing AMI.
Collapse
Affiliation(s)
- Ke Wang
- Department of Clinical Medicine, Xi'an Medical University, Xi'an, Shannxi, China
- Department of the Project of Prevention and Treatment of Respiratory Diseases, Xi'an Medical University, Xi'an, Shannxi, China
| | - Kai Li
- Department of Emergency, Hanjiang Hospital Affiliated to Xi'an Medical University, Hanzhong, Shannxi, China
| | - Zhuoyuan Li
- Department of Clinical Medicine, Xi'an Medical University, Xi'an, Shannxi, China
- Department of the Project of Prevention and Treatment of Respiratory Diseases, Xi'an Medical University, Xi'an, Shannxi, China
| | - Xizhang Yan
- Department of Clinical Medicine, Xi'an Medical University, Xi'an, Shannxi, China
| |
Collapse
|
3
|
Takeda Y, Demura M, Yoneda T, Takeda Y. Epigenetic Regulation of the Renin-Angiotensin-Aldosterone System in Hypertension. Int J Mol Sci 2024; 25:8099. [PMID: 39125667 PMCID: PMC11312206 DOI: 10.3390/ijms25158099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Activation of the renin-angiotensin-aldosterone system (RAAS) plays an important pathophysiological role in hypertension. Increased mRNA levels of the angiotensinogen angiotensin-converting enzyme, angiotensin type 1 receptor gene, Agtr1a, and the aldosterone synthase gene, CYP11B2, have been reported in the heart, blood vessels, and kidneys in salt-sensitive hypertension. However, the mechanism of gene regulation in each component of the RAAS in cardiovascular and renal tissues is unclear. Epigenetic mechanisms, which are important for regulating gene expression, include DNA methylation, histone post-translational modifications, and microRNA (miRNA) regulation. A close association exists between low DNA methylation at CEBP-binding sites and increased AGT expression in visceral adipose tissue and the heart of salt-sensitive hypertensive rats. Several miRNAs influence AGT expression and are associated with cardiovascular diseases. Expression of both ACE and ACE2 genes is regulated by DNA methylation, histone modifications, and miRNAs. Expression of both angiotensinogen and CYP11B2 is reversibly regulated by epigenetic modifications and is related to salt-sensitive hypertension. The mineralocorticoid receptor (MR) exists in cardiovascular and renal tissues, in which many miRNAs influence expression and contribute to the pathogenesis of hypertension. Expression of the 11beta-hydroxysteroid dehydrogenase type 2 (HSD11B2) gene is also regulated by methylation and miRNAs. Epigenetic regulation of renal and vascular HSD11B2 is an important pathogenetic mechanism for salt-sensitive hypertension.
Collapse
Affiliation(s)
- Yoshimichi Takeda
- Endocrinology and Metabolism, Saiseikai Kanazawa Hospital, Kanazawa 920-0353, Japan;
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan;
| | - Masashi Demura
- Department of Hygiene, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan;
| | - Takashi Yoneda
- Institute of Liberal Arts and Science, Kanazawa University, Kanazawa 921-8641, Japan;
- Department of Health Promotion of Medicine of the Future, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan
| | - Yoshiyu Takeda
- Department of Health Promotion of Medicine of the Future, Graduate School of Medical Science, Kanazawa University, Kanazawa 921-8641, Japan
- Hypertension Center, Asanogawa General Hospital, Kanazawa 910-8621, Japan
| |
Collapse
|
4
|
Rezq S, Huffman AM, Basnet J, Alsemeh AE, do Carmo JM, Yanes Cardozo LL, Romero DG. MicroRNA-21 modulates brown adipose tissue adipogenesis and thermogenesis in a mouse model of polycystic ovary syndrome. Biol Sex Differ 2024; 15:53. [PMID: 38987854 PMCID: PMC11238487 DOI: 10.1186/s13293-024-00630-2] [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: 11/18/2022] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with increased obesity, hyperandrogenism, and altered brown adipose tissue (BAT) thermogenesis. MicroRNAs play critical functions in brown adipocyte differentiation and maintenance. We aim to study the role of microRNA-21 (miR-21) in altered energy homeostasis and BAT thermogenesis in a PCOS mouse model of peripubertal androgen exposure. METHODS Three-week-old miR-21 knockout (miR21KO) or wild-type (WT) female mice were treated with dihydrotestosterone (DHT) or vehicle for 90 days. Body composition was determined by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), carbon dioxide production (VCO2), and respiratory exchange ratio (RER) were measured by indirect calorimetry. Androgen receptor (AR), and markers of adipogenesis, de novo lipogenesis, angiogenesis, extracellular matrix remodeling, and thermogenesis were quantified by RT-qPCR and/or Western-blot. RESULTS MiR-21 ablation attenuated DHT-mediated increase in body weight while having no effect on fat or BAT mass. MiR-21 ablation attenuated DHT-mediated BAT AR upregulation. MiR-21 ablation did not alter EE; however, miR21KO DHT-treated mice have reduced VO2, VCO2, and RER. MiR-21 ablation reversed DHT-mediated decrease in food intake and increase in sleep time. MiR-21 ablation decreased some adipogenesis (Adipoq, Pparγ, and Cebpβ) and extracellular matrix remodeling (Mmp-9 and Timp-1) markers expression in DHT-treated mice. MiR-21 ablation abolished DHT-mediated increases in thermogenesis markers Cpt1a and Cpt1b, while decreasing CIDE-A expression. CONCLUSIONS Our findings suggest that BAT miR-21 may play a role in regulating DHT-mediated thermogenic dysfunction in PCOS. Modulation of BAT miR-21 levels could be a novel therapeutic approach for the treatment of PCOS-associated metabolic derangements.
Collapse
Affiliation(s)
- Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| | - Alexandra M Huffman
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Amira E Alsemeh
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Licy L Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Damian G Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
5
|
Tsota M, Giardoglou P, Mentsiou-Nikolaou E, Symianakis P, Kalafati IP, Kyriazopoulou-Korovesi AA, Angelidakis L, Papaioannou M, Konstantaki C, Stamatelopoulos K, Dedoussis GV. Investigation of Antihypertensive Properties of Chios Mastic via Monitoring microRNA-21 Expression Levels in the Plasma of Well-Controlled Hypertensive Patients. Noncoding RNA 2024; 10:33. [PMID: 38921830 PMCID: PMC11207086 DOI: 10.3390/ncrna10030033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Hypertension is a chronic, multifactorial disease, leading to high cardiovascular morbidity and mortality globally. Despite the advantages of pharmaceutical treatments, natural products have gained scientific interest due to their emerging phytotherapeutic properties. Chios mastic is a natural Greek product, consisting of bioactive compounds which modify microRNAs' (small, expression-regulating molecules) expression. In this study, we investigated the antihypertensive properties of Chios mastic through the assessment of miR-21 levels. Herein, plasma samples of 57 individuals with hypertension, recruited for the purposes of the HYPER-MASTIC study, were analyzed. This was a clinical trial with Chios mastic supplements in which the patients were divided into groups receiving high and low mastic doses and placebo supplements, respectively. miR-21 was significantly upregulated in patients compared to normotensive individuals. Mean changes in miR-21 levels were statistically significant, after adjusting for sex and age, between the placebo and low-dose group and between the low- and high-dose group. Post-intervention miR-21 levels were positively associated with night-time systolic blood pressure, pulse pressure, and central systolic mean arterial pressure and negatively associated with night-time pulse wave velocity in the low-dose group. Our findings suggest a potential implication of miR-21 in the association of Chios mastic with night-time blood pressure measurements.
Collapse
Affiliation(s)
- Maria Tsota
- Department of Nutrition-Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece; (M.T.); (E.M.-N.)
| | - Panagiota Giardoglou
- Department of Nutrition-Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece; (M.T.); (E.M.-N.)
| | - Evangelia Mentsiou-Nikolaou
- Department of Nutrition-Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece; (M.T.); (E.M.-N.)
| | - Panagiotis Symianakis
- Department of Nutrition-Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece; (M.T.); (E.M.-N.)
| | - Ioanna Panagiota Kalafati
- Department of Nutrition-Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece; (M.T.); (E.M.-N.)
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-A.K.-K.); (K.S.)
| | | | - Lasthenis Angelidakis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-A.K.-K.); (K.S.)
| | - Maria Papaioannou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-A.K.-K.); (K.S.)
| | - Christina Konstantaki
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-A.K.-K.); (K.S.)
| | - HYPER-MASTIC Consortium
- Department of Nutrition-Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece; (M.T.); (E.M.-N.)
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-A.K.-K.); (K.S.)
- Department of Pharmacy, National and Kapodistrian University of Athens, 15771 Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens, 15772 Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (A.-A.K.-K.); (K.S.)
| | - George V. Dedoussis
- Department of Nutrition-Dietetics, School of Health Science and Education, Harokopio University, 17676 Athens, Greece; (M.T.); (E.M.-N.)
| |
Collapse
|
6
|
Wang X, Wu H, An J, Zhang G, Chen Y, Fu L, Tao L, Liang G, Shen X. Cyclovirobuxine D alleviates aldosterone-induced myocardial hypertrophy by protecting mitochondrial function depending on the mutual regulation of Nrf2-SIRT3. Biomed Pharmacother 2023; 167:115618. [PMID: 37793277 DOI: 10.1016/j.biopha.2023.115618] [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: 07/24/2023] [Revised: 09/24/2023] [Accepted: 09/28/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Cyclovirobuxine D (CVB-D) is a natural alkaloid that exhibits multiple pharmacological activities, such as anti-inflammatory, anti-oxidative stress, and anti-cancer properties. However, its specific protective mechanism of action for myocardial hypertrophy remains unresolved. PURPOSE This work was to investigate the ameliorative impact of CVB-D in myocardial hypertrophy, and to elucidate aldosterone (ALD)-induced myocardial hypertrophy by inhibiting the SIRT3 mediated Nrf2 activation. METHODS The myocardial hypertrophy model was reproduced by ALD both in vitro and in vivo, and the protective effect of CVB-D on myocardium and mitochondria was evaluated by TEM, H&E, qPCR, Western blot and ChIP. An immunoprecipitation experiment was adopted to evaluate the acetylation level of Nrf2 and the binding between SIRT3 and Nrf2. Additionally, bardoxolone-methyl (BAR, an Nrf2 agonist), ML385 (an Nrf2 inhibitor), resveratrol (RES, a SIRT3 agonist), and 3-TYP (a SIRT3 inhibitor) were used to confirm the molecular mechanism of CVB-D. Lastly, a molecular docking technique was employed to predict the binding site of SIRT3 and Nrf2 proteins. RESULTS Our findings suggested that CVB-D improved mitochondrial function, leading to a reduction in ALD-induced cardiomyocyte hypertrophy. By CVB-D treatment, there was an activation of mutual regulation between Nrf2 and SIRT3. Specifically, CVB-D resulted in the increase of Nrf2 protein in the nucleus and activated Nrf2 signaling pathway, thus up-regulating SIRT3. The activation of SIRT3 and the protective action of mitochondrion disappeared because of the intervention of ML385. After CVB-D activated SIRT3, the acetylation level of Nrf2 decreased, followed by activating the Nrf2 pathway. The activation of Nrf2 and mitochondrial protection by CVB-D were reversed by 3-TYP. Our results are also supported by Co-IP and molecular docking analysis, revealing that CVB-D promotes SIRT3-mediated Nrf2 activation. CONCLUSION Thus, CVB-D ameliorates ALD-induced myocardial hypertrophy by recovering mitochondrial function by activating the mutual regulation of Nrf2 and SIRT3. Thus, CVB-D could be a beneficial drug for myocardial hypertrophy.
Collapse
Affiliation(s)
- Xueting Wang
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China; Medical College, Guizhou University, Huaxi District, 550025 Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guian New District, 550025 Guizhou, China
| | - Hongkun Wu
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China
| | - Jiangfei An
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guian New District, 550025 Guizhou, China
| | - Guangqiong Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guian New District, 550025 Guizhou, China
| | - Yan Chen
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guian New District, 550025 Guizhou, China
| | - Lingyun Fu
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guian New District, 550025 Guizhou, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guian New District, 550025 Guizhou, China
| | - Guiyou Liang
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China.
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants (The Key Laboratory of Endemic and Ethnic Diseases of Ministry of Education), Guizhou Medical University, Guian New District, 550025 Guizhou, China; The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province and The high educational key laboratory of Guizhou province for natural medicianl Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, Guian New District, 550025 Guizhou, China.
| |
Collapse
|
7
|
MacKenzie SM, Birch LA, Lamprou S, Rezvanisanijouybari P, Fayad M, Zennaro MC, Davies E. MicroRNAs in aldosterone production and action. VITAMINS AND HORMONES 2023; 124:137-163. [PMID: 38408798 DOI: 10.1016/bs.vh.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Aldosterone is a cardiovascular hormone with a key role in blood pressure regulation, among other processes, mediated through its targeting of the mineralocorticoid receptor in the renal tubule and selected other tissues. Its secretion from the adrenal gland is a highly controlled process subject to regulatory influence from the renin-angiotensin system and the hypothalamic-pituitary-adrenal axis. MicroRNAs are small endogenous non-coding RNA molecules capable of regulating gene expression post-transcriptionally through stimulation of mRNA degradation or suppression of translation. Several studies have now identified that microRNA levels are changed in cases of aldosterone dysregulation and that microRNAs are capable of regulating the expression of various genes involved in aldosterone production and action. In this article we summarise the major studies concerning this topic. We also discuss the potential role for circulating microRNAs as diagnostic biomarkers for primary aldosteronism, a highly treatable form of secondary hypertension, which would be highly desirable given the current underdiagnosis of this condition.
Collapse
Affiliation(s)
- Scott M MacKenzie
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom.
| | - Lara A Birch
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Stelios Lamprou
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Parisa Rezvanisanijouybari
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - May Fayad
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom; Université Paris Cité, PARCC, INSERM, Paris, France
| | - Maria-Christina Zennaro
- Université Paris Cité, PARCC, INSERM, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Eleanor Davies
- School of Cardiovascular and Metabolic Health, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
8
|
Wu M, Li G, Wang W, Ren H. Emerging roles of microRNAs in septic cardiomyopathy. Front Pharmacol 2023; 14:1181372. [PMID: 37475718 PMCID: PMC10354437 DOI: 10.3389/fphar.2023.1181372] [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: 03/07/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023] Open
Abstract
As one of the serious complications of sepsis, septic cardiomyopathy has gained more and more attention, because of its high morbidity and mortality. With the in-depth study of septic cardiomyopathy, several methods have been adopted clinically but have poor therapeutic effects due to failure to find precise therapeutic targets. In recent years, microRNAs have been found to be related to the pathogenesis, diagnosis, and treatment of septic cardiomyopathy via regulating immunity and programmed cell death. This paper reviews the role of microRNAs in septic cardiomyopathy, aiming to provide new targets for the diagnosis and treatment of septic cardiomyopathy.
Collapse
Affiliation(s)
| | | | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hongsheng Ren
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
9
|
Tapia-Castillo A, Carvajal CA, Pérez JA, Fardella CE. Clinical, biochemical, and miRNA profile of subjects with positive screening of primary aldosteronism and nonclassic apparent mineralocorticoid excess. Endocrine 2022; 77:380-391. [PMID: 35676467 DOI: 10.1007/s12020-022-03103-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/27/2022] [Indexed: 12/15/2022]
Abstract
Primary aldosteronism (PA) and nonclassic apparent mineralocorticoid excess (NCAME) have been recognized as endocrine-related conditions having a broad clinical-biochemical spectrum, spanning from normotension to severe arterial hypertension (AHT). However, the coexistence of both phenotypes have not been reported to date. AIM To identify and characterize clinical and biochemical parameters of subjects with both PA and NCAME conditions (NCAME&PA) and study the miRNA cargo in their urinary extracellular vesicles as potential biomarkers for this novel condition. METHODS We performed a cross-sectional study of 206 Chilean adult subjects from a primary care cohort. We measured blood pressure (BP), cortisol (F), cortisone (E), aldosterone, plasma renin activity (PRA), microalbuminuria (MAC), plasma NGAL, MMP9, fractional-potassium-excretion (FEK). Subjects were classified as NCAME&PA, PA, NCAME, essential hypertensives (EH), or healthy controls (CTL). EV-miRNAs were quantified by Taqman-qPCR. RESULTS We found that 30.6% subjects had an abnormal endocrine phenotype: NCAME&PA (6.8%), PA (11.2%) or NCAME (12.6%), and the prevalence of AHT was 92.9%, 82.6%, and 65%, respectively. NCAME&PA subjects had both lower cortisone (p < 0.05) and lower PRA (p < 0.0001), higher FEK (p = 0.02) and higher MAC (p = 0.01) than EH or CTL. NCAME&PA subjects had also higher NGAL levels than CTL and PA (p < 0.05). Exosome miR-192, miR-133a and miR-21 expression decreased with phenotype severity and correlated with BP and PRA (p < 0.05). CONCLUSION We identified adult subjects with a combined condition of NCAME and PA associated with higher BP, increased renal and endothelial damage markers than control and EH. Additionally, we observed a differential expression of a specific miRNAs, suggesting a potential role of these miRNAs associated to this novel combined phenotype.
Collapse
Affiliation(s)
- Alejandra Tapia-Castillo
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy (IMII-ICM), Santiago, Chile
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian A Carvajal
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy (IMII-ICM), Santiago, Chile
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Pérez
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy (IMII-ICM), Santiago, Chile
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos E Fardella
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
10
|
Li H, Zhan J, Chen C, Wang D. MicroRNAs in cardiovascular diseases. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:140-168. [PMID: 37724243 PMCID: PMC10471109 DOI: 10.1515/mr-2021-0001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 12/29/2021] [Indexed: 09/20/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death and disability worldwide, despite the wide diversity of molecular targets identified and the development of therapeutic methods. MicroRNAs (miRNAs) are a class of small (about 22 nucleotides) non-coding RNAs (ncRNAs) that negatively regulate gene expression at the post-transcriptional level in the cytoplasm and play complicated roles in different CVDs. While miRNA overexpression in one type of cell protects against heart disease, it promotes cardiac dysfunction in another type of cardiac cell. Moreover, recent studies have shown that, apart from cytosolic miRNAs, subcellular miRNAs such as mitochondria- and nucleus-localized miRNAs are dysregulated in CVDs. However, the functional properties of cellular- and subcellular-localized miRNAs have not been well characterized. In this review article, by carefully revisiting animal-based miRNA studies in CVDs, we will address the regulation and functional properties of miRNAs in various CVDs. Specifically, the cell-cell crosstalk and subcellular perspective of miRNAs are highlighted. We will provide the background for attractive molecular targets that might be useful in preventing the progression of CVDs and heart failure (HF) as well as insights for future studies.
Collapse
Affiliation(s)
- Huaping Li
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiabing Zhan
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| |
Collapse
|
11
|
Carvajal CA, Tapia-Castillo A, Pérez JA, Fardella CE. Primary Aldosteronism, Aldosterone, and Extracellular Vesicles. Endocrinology 2022; 163:6433012. [PMID: 34918071 DOI: 10.1210/endocr/bqab240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Indexed: 01/02/2023]
Abstract
Primary aldosteronism (PA) is an endocrine related condition leading to arterial hypertension due to inappropriately high and unregulated aldosterone concentration. Recently, a broad spectrum of PA has been recognized, which brings new challenges associated with early identification of this condition that affect renal epithelial and extrarenal tissues. Reports have shown the potential role of extracellular vesicles (EVs) and EV cargo as novel and complementary biomarkers in diagnosis and prognosis of PA. In vivo and in vitro studies have identified specific EV surface antigens, EV-proteins, and EV microRNAs that can be useful to develop novel diagnostic algorithms to detect, confirm, or follow up the PA. Moreover, the study of EVs in the field of PA provides further insight in the pathophysiological mechanism of the PA disease.
Collapse
Affiliation(s)
- Cristian A Carvajal
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Tapia-Castillo
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Pérez
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos E Fardella
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile
- Centro Traslacional de Endocrinología UC (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
12
|
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: 114] [Impact Index Per Article: 38.0] [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
|
13
|
Improta-Caria AC, Aras MG, Nascimento L, De Sousa RAL, Aras-Júnior R, Souza BSDF. MicroRNAs Regulating Renin-Angiotensin-Aldosterone System, Sympathetic Nervous System and Left Ventricular Hypertrophy in Systemic Arterial Hypertension. Biomolecules 2021; 11:biom11121771. [PMID: 34944415 PMCID: PMC8698399 DOI: 10.3390/biom11121771] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs are small non-coding RNAs that regulate gene and protein expression. MicroRNAs also regulate several cellular processes such as proliferation, differentiation, cell cycle, apoptosis, among others. In this context, they play important roles in the human body and in the pathogenesis of diseases such as cancer, diabetes, obesity and hypertension. In hypertension, microRNAs act on the renin-angiotensin-aldosterone system, sympathetic nervous system and left ventricular hypertrophy, however the signaling pathways that interact in these processes and are regulated by microRNAs inducing hypertension and the worsening of the disease still need to be elucidated. Thus, the aim of this review is to analyze the pattern of expression of microRNAs in these processes and the possible associated signaling pathways.
Collapse
Affiliation(s)
- Alex Cleber Improta-Caria
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Salvador 40110-100, Brazil;
- Department of Physical Education in Cardiology of the State of Bahia, Brazilian Society of Cardiology, Salvador 41170-130, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador 41253-190, Brazil
- Correspondence: (A.C.I.-C.); (B.S.d.F.S.)
| | - Marcela Gordilho Aras
- Faculty of Medicine, Federal University of Bahia, Salvador 40110-100, Brazil; (M.G.A.); (L.N.)
| | - Luca Nascimento
- Faculty of Medicine, Federal University of Bahia, Salvador 40110-100, Brazil; (M.G.A.); (L.N.)
| | | | - Roque Aras-Júnior
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Salvador 40110-100, Brazil;
- Faculty of Medicine, Federal University of Bahia, Salvador 40110-100, Brazil; (M.G.A.); (L.N.)
| | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador 41253-190, Brazil
- D’Or Institute for Research and Education (IDOR), Salvador 22281-100, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador 40296-710, Brazil
- Correspondence: (A.C.I.-C.); (B.S.d.F.S.)
| |
Collapse
|
14
|
Carvajal CA, Tapia-Castillo A, Pérez JA, Fardella CE. Serum Alpha-1-Acid Glycoprotein-1 and Urinary Extracellular Vesicle miR-21-5p as Potential Biomarkers of Primary Aldosteronism. Front Immunol 2021; 12:768734. [PMID: 34804057 PMCID: PMC8603108 DOI: 10.3389/fimmu.2021.768734] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
Primary aldosteronism (PA) is the most common cause of secondary hypertension and reaches a prevalence of 6-10%. PA is an endocrine disorder, currently identified as a broad-spectrum phenotype, spanning from normotension to hypertension. In this regard, several studies have made advances in the identification of mediators and novel biomarkers of PA as specific proteins, miRNAs, and lately, extracellular vesicles (EVs) and their cargo. Aim To evaluate lipocalins LCN2 and AGP1, and specific urinary EV miR-21-5p and Let-7i-5p as novel biomarkers for PA. Subjects and Methods A cross-sectional study was performed in 41 adult subjects classified as normotensive controls (CTL), essential hypertensives (EH), and primary aldosteronism (PA) subjects, who were similar in gender, age, and BMI. Systolic (SBP) and diastolic (DBP) blood pressure, aldosterone, plasma renin activity (PRA), and aldosterone to renin ratio (ARR) were determined. Inflammatory parameters were defined as hs-C-reactive protein (hs-CRP), PAI-1, MMP9, IL6, LCN2, LCN2-MMP9, and AGP1. We isolated urinary EVs (uEVs) and measured two miRNA cargo miR-21-5p and Let-7i-5p by Taqman-qPCR. Statistical analyses as group comparisons were performed by Kruskall-Wallis, and discriminatory analyses by ROC curves were performed with SPSS v21 and Graphpad-Prism v9. Results PA and EH subjects have significantly higher SBP and DBP (p <0.05) than the control group. PA subjects have similar hs-CRP, PAI-1, IL-6, MMP9, LCN2, and LCN2-MMP9 but have higher levels of AGP1 (p <0.05) than the CTL&EH group. The concentration and size of uEVs and miRNA Let-7i-5p did not show any difference between groups. In PA, we found significantly lower levels of miR-21-5p than controls (p <0.05). AGP1 was associated with aldosterone, PRA, and ARR. ROC curves detected AUC for AGP1 of 0.90 (IC 95 [0.79 - 1.00], p <0.001), and combination of AGP1 and EV-miR-21-5p showed an AUC of 0.94 (IC 95 [0.85 - 1.00], p<0.001) to discriminate the PA condition from EH and controls. Conclusion Serum AGP1 protein was found to be increased, and miR-21-5p in uEVs was decreased in subjects classified as PA. Association of AGP1 with aldosterone, renin activity, and ARR, besides the high discriminatory capacity of AGP1 and uEV-miR-21-5p to identify the PA condition, place both as potential biomarkers of PA.
Collapse
Affiliation(s)
- Cristian A Carvajal
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Tapia-Castillo
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Pérez
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos E Fardella
- Department of Endocrinology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Endocrinology, Millennium Institute of Immunology and Immunotherapy (IMII-ICM), Santiago, Chile.,Center for Translational Research in Endocrinology (CETREN-UC), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
15
|
Rezq S, Huffman AM, Basnet J, Yanes Cardozo LL, Romero DG. Cardiac and Renal SARS-CoV-2 Viral Entry Protein Regulation by Androgens and Diet: Implications for Polycystic Ovary Syndrome and COVID-19. Int J Mol Sci 2021; 22:ijms22189746. [PMID: 34575910 PMCID: PMC8470275 DOI: 10.3390/ijms22189746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 12/31/2022] Open
Abstract
The susceptibility and the severity of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are associated with hyperandrogenism, obesity, and preexisting pulmonary, metabolic, renal, and cardiac conditions. Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with obesity, hyperandrogenism, and cardiometabolic dysregulations. We analyzed cardiac, renal, circulatory, and urinary SARS-CoV-2 viral entry proteins (ACE2, TMPRSS2, TMPRSS4, furin, cathepsin L, and ADAM17) and androgen receptor (AR) expression, in a peripubertal androgen exposure model of PCOS. Peripubertal female mice were treated with dihydrotestosterone (DHT) and low (LFD) or high (HFD) fat diet for 90 days. HFD exacerbated DHT-induced increase in body weight, fat mass, and cardiac and renal hypertrophy. In the heart, DHT upregulated AR protein in both LFD and HFD, ACE2 in HFD, and ADAM17 in LFD. In the kidney, AR protein expression was upregulated by both DHT and HFD. Moreover, ACE2 and ADAM17 were upregulated by DHT in both diets. Renal TMPRSS2, furin, and cathepsin L were upregulated by DHT and differentially modulated by the diet. DHT upregulated urinary ACE2 in both diets, while neither treatment modified serum ACE2. Renal AR mRNA expression positively correlated with Ace2, Tmprss2, furin, cathepsin L, and ADAM17. Our findings suggest that women with PCOS could be a population with a high risk of COVID-19-associated cardiac and renal complications. Furthermore, our study suggests that weight loss by lifestyle modifications (i.e., diet) could potentially mitigate COVID-19-associated deleterious cardiorenal outcomes in women with PCOS.
Collapse
Affiliation(s)
- Samar Rezq
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA; (S.R.); (A.M.H.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Alexandra M. Huffman
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA; (S.R.); (A.M.H.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
| | - Jelina Basnet
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA; (S.R.); (A.M.H.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
| | - Licy L. Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA; (S.R.); (A.M.H.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
| | - Damian G. Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA; (S.R.); (A.M.H.); (J.B.); (L.L.Y.C.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, 2500 N, State Street, Jackson, MS 39216, USA
- Correspondence: ; Tel.: +1-601-984-1523; Fax: +1-601-984-1501
| |
Collapse
|
16
|
Bai L, Zhao Y, Zhao L, Zhang M, Cai Z, Yung KKL, Dong C, Li R. Ambient air PM 2.5 exposure induces heart injury and cardiac hypertrophy in rats through regulation of miR-208a/b, α/β-MHC, and GATA4. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 85:103653. [PMID: 33812011 DOI: 10.1016/j.etap.2021.103653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 06/12/2023]
Abstract
Ambient air fine particulate matter (PM2.5) may increase cardiovascular disease risks. In this study, we investigated the miR-208/GATA4/myosin heavy chain (MHC) regulation mechanisms on cardiac injury in rats after PM2.5 exposure via an animal inhalation device. The results showed that PM2.5 exposure for 2 months caused pathological heart injury, reduced nucleus-cytoplasm ratio, and increased the levels of CK-MB and cTnI, showing cardiac hypertrophy. Oxidative stress and inflammatory responses were also observed in rats' hearts exposed to PM2.5. Of note, PM2.5 exposure for 2-month significantly elevated GATA4 and β-MHC mRNA and protein expression compared with the corresponding controls, along with the high-expression of miR-208b. The ratios of β-MHC/α-MHC expression induced by PM2.5 were remarkably raised in comparison to their controls. It suggested that the up-regulation of miR-208b/β-MHC and GATA4 and the conversion from α-MHC to β-MHC may be the important causes of cardiac hypertrophy in rats incurred by PM2.5.
Collapse
Affiliation(s)
- Lirong Bai
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Yufei Zhao
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Lifang Zhao
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Mei Zhang
- Institute of Environmental Science, Shanxi University, Taiyuan, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Ken Kin Lam Yung
- Institute of Environmental Science, Shanxi University, Taiyuan, China; Department of Biology, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan, China.
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, China.
| |
Collapse
|
17
|
Zhang J, Liu Y, Liu L. Hyperoside prevents sepsis-associated cardiac dysfunction through regulating cardiomyocyte viability and inflammation via inhibiting miR-21. Biomed Pharmacother 2021; 138:111524. [PMID: 34311527 DOI: 10.1016/j.biopha.2021.111524] [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: 12/16/2020] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Sepsis-associated cardiac dysfunction results in increased mortality. Hyperoside (Hyp) is a flavonoid, showing significant anti-inflammatory effects. However, its pharmacological effects on sepsis-induced cardiac dysfunction remain unknown. In this study, we attempted to explore whether Hyp could prevent cardiac dysfunction and its underlying mechanisms. METHODS We established a mice mode of sepsis by cecal ligation and puncture (CLP) treatment, and constructed a cell model of myocardial injury by lipopolysaccharide (LPS) stimulation. The cardiac function indicators and the inflammatory cytokine levels were measured. Effect of Hyp on cardiomyocyte viability was evaluated using MTT assay. The expression and functional role of microRNA-21 (miR-21), a documented molecule that regulated by Hyp, was evaluated in the constructed models, and the potential targets of miR-21 were predicted. RESULTS Hyp alleviated the impaired cardiac function and stimulated inflammation caused by CLP in the in vivo sepsis model, and alleviated the LPS-induced decrease in cell viability and increase in inflammation of cardiomyocytes. Additionally, Hyp significantly inhibited the expression of miR-21 in LPS-induced cardiomyocytes, and the increased cell viability and decreased inflammation caused by Hyp in the in vitro model could be reversed by miR-21 overexpression. In animal model of sepsis, the protective influence of Hyp against sepsis-induced cardiac dysfunction was attenuated by miR-21 upregulation. CONCLUSION Our findings demonstrated that Hyp may serve as a promising natural drug for the treatment of sepsis-associated cardiac dysfunction, and its protective role may exerted through regulating cardiomyocyte viability and inflammation by suppressing miR-21.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Pharmacy, Weifang Maternal and Child Health Hospital, Weifang 261011, Shandong, China
| | - Yujuan Liu
- Central Supply Room, Weifang Maternal and Child Health Hospital, Weifang 261011, Shandong, China
| | - Lijun Liu
- Department of Gynecology, Weifang Maternal and Child Health Hospital, Weifang 261011, Shandong, China.
| |
Collapse
|
18
|
Pruett JE, Torres Fernandez ED, Everman SJ, Vinson RM, Davenport K, Logan MK, Ye SA, Romero DG, Yanes Cardozo LL. Impact of SGLT-2 Inhibition on Cardiometabolic Abnormalities in a Rat Model of Polycystic Ovary Syndrome. Int J Mol Sci 2021; 22:2576. [PMID: 33806551 PMCID: PMC7962009 DOI: 10.3390/ijms22052576] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/21/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder in reproductive-age women. PCOS is characterized by hyperandrogenism and ovulatory dysfunction. Women with PCOS have a high prevalence of obesity, insulin resistance (IR), increased blood pressure (BP), and activation of the renin angiotensin system (RAS). Effective evidence-based therapeutics to ameliorate the cardiometabolic complications in PCOS are lacking. The sodium-glucose cotransporter-2 (SGLT2) inhibitor Empagliflozin (EMPA) reduces BP and hyperglycemia in type 2 diabetes mellitus. We hypothesized that hyperandrogenemia upregulates renal SGLT2 expression and that EMPA ameliorates cardiometabolic complications in a hyperandrogenemic PCOS model. Four-week-old female Sprague Dawley rats were treated with dihydrotestosterone (DHT) for 90 days, and EMPA was co-administered for the last three weeks. DHT upregulated renal SGLT2, SGLT4, and GLUT2, but downregulated SGLT3 mRNA expression. EMPA decreased DHT-mediated increases in fat mass, plasma leptin, and BP, but failed to decrease plasma insulin, HbA1c, or albuminuria. EMPA decreased DHT-mediated increase in renal angiotensin converting enzyme (ACE), angiotensin converting enzyme 2 (ACE2), and angiotensin II type 1 receptor (AGT1R) mRNA and protein expression. In summary, SGLT2 inhibition proved beneficial in adiposity and BP reduction in a hyperandrogenemic PCOS model; however, additional therapies may be needed to improve IR and renal injury.
Collapse
Affiliation(s)
- Jacob E. Pruett
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
| | - Edgar D. Torres Fernandez
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
| | - Steven J. Everman
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
| | - Ruth M. Vinson
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
| | - Kacey Davenport
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
| | - Madelyn K. Logan
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
| | - Stephanie A. Ye
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
| | - Damian G. Romero
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Licy L. Yanes Cardozo
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA; (J.E.P.); (E.D.T.F.); (S.J.E.); (R.M.V.); (K.D.); (M.K.L.); (S.A.Y.); (D.G.R.)
- Mississippi Center of Excellence in Perinatal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Women’s Health Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Cardio Renal Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Division of Endocrinology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
19
|
Geng H, Su Y, Huang R, Fan M, Li X, Lu X, Sheng H. Specific protein 1 inhibitor mithramycin A protects cardiomyocytes from myocardial infarction via interacting with PARP. In Vitro Cell Dev Biol Anim 2021; 57:315-323. [PMID: 33580416 DOI: 10.1007/s11626-021-00543-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Specific protein 1 (SP1) might act as a critical transcription regulator in myocardial infarction (MI), but little evidence about its function in regulating cardiac apoptosis, a major cause of MI development, has been revealed. This study tried to investigate the role of SP1 in MI and its interaction with poly-ADP-ribose polymerase (PARP)-1 by using SP1 inhibitor, mithramycin A (mithA). Primary mouse cardiomyocytes and commercial mouse cardiomyocytes were subjected to mithA treatment under hypoxia conditions, while cell viability, Nix promoter activity, and its expression were detected correspondingly. PARP overexpression and knockdown were conducted, respectively, in mithA-treated and SP1-overexpressing cells. Co-immunoprecipitation was used to verify the interaction between PARP and SP1. For in vivo experiments, mithA administration was performed after the injections of adenovirus for PARP overexpression, and then, MI introduction was carried out. Infarct size and lactate dehydrogenase level were measured to assess MI injury. SP1 inhibitor mithA attenuated hypoxia-induced decrease of cell viability and Nix transcriptional activation, which could be inhibited by PARP overexpression. Knockdown of PARP prevented SP1-induced transcription of Nix and cell viability change, and PARP showed direct interaction with SP1. Furthermore, mithA administration reduced MI injuries, while PARP overexpression could suppress the improvement. The cardioprotective role of SP1 inhibitor mithA was demonstrated here expanding the role of SP1 in MI development involving hypoxia-induced cardiac apoptosis. Moreover, PARP acted as a transcriptional coactivator in Nix transcription involving its interaction with SP1.
Collapse
Affiliation(s)
- Haihua Geng
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Yamin Su
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Rong Huang
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Mengkang Fan
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Xiaofei Li
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Xiaochen Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China
| | - Hongzhuan Sheng
- Department of Cardiology, Affiliated Hospital of Nantong University, No. 20 Xisi Rd, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
20
|
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: 136] [Impact Index Per Article: 34.0] [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
|
21
|
Li X, Yang Y, Chen S, Zhou J, Li J, Cheng Y. Epigenetics-based therapeutics for myocardial fibrosis. Life Sci 2021; 271:119186. [PMID: 33577852 DOI: 10.1016/j.lfs.2021.119186] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Myocardial fibrosis (MF) is a reactive remodeling process in response to myocardial injury. It is mainly manifested by the proliferation of cardiac muscle fibroblasts and secreting extracellular matrix (ECM) proteins to replace damaged tissue. However, the excessive production and deposition of extracellular matrix, and the rising proportion of type I and type III collagen lead to pathological fibrotic remodeling, thereby facilitating the development of cardiac dysfunction and eventually causing heart failure with heightened mortality. Currently, the molecular mechanisms of MF are still not fully understood. With the development of epigenetics, it is found that epigenetics controls the transcription of pro-fibrotic genes in MF by DNA methylation, histone modification and noncoding RNAs. In this review, we summarize and discuss the research progress of the mechanisms underlying MF from the perspective of epigenetics, including the newest m6A modification and crosstalk between different epigenetics in MF. We also offer a succinct overview of promising molecules targeting epigenetic regulators, which may provide novel therapeutic strategies against MF.
Collapse
Affiliation(s)
- Xuping Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Ying Yang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Sixuan Chen
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jiuyao Zhou
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jingyan Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Yuanyuan Cheng
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
22
|
TRIF/miR-34a mediates aldosterone-induced cardiac inflammation and remodeling. Clin Sci (Lond) 2020; 134:1319-1331. [PMID: 32542395 DOI: 10.1042/cs20200249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/23/2022]
Abstract
Aldosterone, as a major product of renin-angiotensin-aldosterone system (RAAS), determines multiple pathophysiological processes in cardiovascular diseases. The excess inflammatory response is one of the key profiles in aldosterone-mediated cardiac remodeling. However, the potential mechanisms of aldosterone/inflammatory signaling were still not fully disclosed. The present study aimed to investigate whether TIR-domain-containing adapter-inducing interferon-β (Trif) participated in the aldosterone-induced cardiac remodeling, and to explore potential molecular mechanisms. Trif knockout mice and their littermates were osmotically administrated with aldosterone (50 μg/kg per day) for 21 and 42 days. The cardiac structural analysis, functional parameters, and mitochondrial function were measured. Aldosterone dose- or time-dependently increased the levels of TRIF in primary mouse cardiomyocytes or mouse heart tissues. Trif deficiency protected against aldosterone-induced cardiac hypertrophy, fibrosis and dysfunction. Moreover, Trif deficiency also suppressed aldosterone-induced cardiac inflammatory response and mitochondrial injuries. Mechanistically, overexpression of cardiac microRNAs (miR)-34a reversed the cardiac benefits of Trif deficiency in aldosterone-treated mice. Taken together, Trif/miR-34a axis could provide a novel molecular mechanism for explaining aldosterone-induced cardiac hypertrophy, fibrosis and functional disorders.
Collapse
|
23
|
Ben-Nun D, Buja LM, Fuentes F. Prevention of heart failure with preserved ejection fraction (HFpEF): reexamining microRNA-21 inhibition in the era of oligonucleotide-based therapeutics. Cardiovasc Pathol 2020; 49:107243. [PMID: 32629211 DOI: 10.1016/j.carpath.2020.107243] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for 50% of cases of heart failure, which is the most common cause of hospitalization in US patients over the age of 65. HFpEF pathogenesis is increasingly believed to be due to pathological hypertrophy and fibrosis of the myocardium that may be a result of systemic inflammation from comorbid conditions such as hypertension, diabetes mellitus, chronic obstructive pulmonary disease, anemia, chronic kidney disease and others. It is believed that oxidative stress triggers a process of pathological hypertrophy and fibrosis in cardiac endothelial cells, which leads to increased left ventricle filling pressures and, eventually, symptoms of heart failure. Numerous recent major clinical trials that have examined various therapies aimed at improving mortality in HFpEF have emerged empty-handed and thus the search for effective management strategies continues. Over the last several years, there have been many new developments in the field of antisense oligonucleotide-based therapeutics, which involves using noncoding nucleic acid particles such as microRNA and small interfering RNA to repress the expression of specific messenger RNA. In this article, we review the concept of using oligonucleotide-based therapeutics to prevent or treat HFpEF by targeting a specific microRNA that has been implicated in the pathogenesis of myocardial fibrosis and hypertrophy, microRNA-21 (miR-21). We review the various evidence that implicates miR-21 in the process of myocardial fibrosis and discuss recent attempts to use antimiR-21 compounds to prevent fibrosis. We also discuss proposed methods for screening patients at high risk for HFpEF for diastolic dysfunction in order to determine which patients.
Collapse
Affiliation(s)
- David Ben-Nun
- Tel Aviv University Sackler Faculty of Medicine, Sackler Faculty of Medicine, NY St..., 69978 Tel Aviv, Israel.
| | - L Maximilian Buja
- The University of Texas Health Science Center at Houston, McGovern Medical School
| | - Francisco Fuentes
- The University of Texas Health Science Center at Houston, McGovern Medical School
| |
Collapse
|
24
|
AlQudah M, Hale TM, Czubryt MP. Targeting the renin-angiotensin-aldosterone system in fibrosis. Matrix Biol 2020; 91-92:92-108. [PMID: 32422329 DOI: 10.1016/j.matbio.2020.04.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
Abstract
Fibrosis is characterized by excessive deposition of extracellular matrix components such as collagen in tissues or organs. Fibrosis can develop in the heart, kidneys, liver, skin or any other body organ in response to injury or maladaptive reparative processes, reducing overall function and leading eventually to organ failure. A variety of cellular and molecular signaling mechanisms are involved in the pathogenesis of fibrosis. The renin-angiotensin-aldosterone system (RAAS) interacts with the potent Transforming Growth Factor β (TGFβ) pro-fibrotic pathway to mediate fibrosis in many cell and tissue types. RAAS consists of both classical and alternative pathways, which act to potentiate or antagonize fibrotic signaling mechanisms, respectively. This review provides an overview of recent literature describing the roles of RAAS in the pathogenesis of fibrosis, particularly in the liver, heart, kidney and skin, and with a focus on RAAS interactions with TGFβ signaling. Targeting RAAS to combat fibrosis represents a promising therapeutic approach, particularly given the lack of strategies for treating fibrosis as its own entity, thus animal and clinical studies to examine the impact of natural and synthetic substances to alter RAAS signaling as a means to treat fibrosis are reviewed as well.
Collapse
Affiliation(s)
- Mohammad AlQudah
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Canada; Department of Physiology and Biochemistry, College of Medicine, Jordan University of Science and Technology, Jordan
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona College of Medicine Phoenix, United States
| | - Michael P Czubryt
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Canada.
| |
Collapse
|
25
|
Chen H, Xu Y, Rappold A, Diaz-Sanchez D, Tong H. Effects of ambient ozone exposure on circulating extracellular vehicle microRNA levels in coronary artery disease patients. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2020; 83:351-362. [PMID: 32414303 PMCID: PMC7306136 DOI: 10.1080/15287394.2020.1762814] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Exposure to ambient air pollutants such as ozone (O3) and particulate matter (PM) is associated with increased cardiovascular morbidity and rate of mortality, but the underlying biological mechanisms have yet to be described. Emerging evidence shows that extracellular vehicle (EV) microRNAs (miRNAs) may facilitate cell-to-cell and organ-to-organ communications and play a role in the air pollution-induced cardiovascular effects. This study aims to explore the association between air pollutant exposure and miRNA changes related to cardiovascular diseases. Using a panel study design, 14 participants with coronary artery diseases were enrolled in this study. Each participant had up to 10 clinical visits and their plasma samples were collected and measured for expression of miRNA-21 (miR-21), miR-126, miR-146, miR-150, and miR-155. Mixed effects models adjusted for temperature, humidity, and season were used to examine the association between miRNA levels and exposure to 8-hr O3 or 24-hr PM2.5 up to 4 days prior. Results demonstrated that miR-150 expression was positively associated with O3 exposure at 1-4 days lag and 5day moving average while miR-155 expression tracked with O3 exposure at lag 0. No significant association was found between miRNA expression and ambient PM2.5 at any time point. β-blocker and diabetic medication usage significantly modified the correlation between O3 exposure and miR-150 expression where the link was more prominent among non-users. In conclusion, evidence indicated an association between exposure to ambient O3 and circulating levels of EV miR-150 and miR-155 was observed. These findings pointed to a future research direction involving miRNA-mediated mechanisms of O3-induced cardiovascular effects.
Collapse
Affiliation(s)
- Hao Chen
- Oak Ridge Institute of Science and Education, 100 ORAU Way, Oak Ridge, TN 37830, USA
| | - Yunan Xu
- Department of Psychiatry and Behavioral Sciences, Duke University, 905 W. Main Street, Durham, NC 27701, USA
| | - Ana Rappold
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC 27514, USA
| | - David Diaz-Sanchez
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC 27514, USA
| | - Haiyan Tong
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, 104 Mason Farm Road, Chapel Hill, NC 27514, USA
| |
Collapse
|
26
|
The association between microRNA-21 and hypertension-induced cardiac remodeling. PLoS One 2020; 15:e0226053. [PMID: 32040481 PMCID: PMC7010249 DOI: 10.1371/journal.pone.0226053] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Hypertension is a major public health problem among the aging population worldwide. It causes cardiac remodeling, including hypertrophy and interstitial fibrosis, which leads to development of hypertensive heart disease (HHD). Although microRNA-21 (miR-21) is associated with fibrogenesis in multiple organs, its contribution to cardiac remodeling in hypertension is poorly understood. Circulating miR-21 level was higher in patients with HHD than that in the control subjects. It also positively correlated with serum myocardial fibrotic markers. MiR-21 expression levels were significantly upregulated in the mice hearts after angiotensin II (Ang II) infusion or transverse aortic constriction (TAC) compared with control mice. Expression level of programmed cell death 4 (PDCD4), a main target of miR-21, was significantly decreased in Ang II infused mice and TAC mice compared with control mice. Expression levels of transcriptional activator protein 1 (AP-1) and transforming growth factor-β1 (TGF-β1), which were downstream targets of PDCD4, were increased in Ang II infused mice and TAC mice compared with control mice. In vitro, mirVana-miR-21-specific inhibitor attenuated Ang II-induced PDCD4 downregulation and contributed to subsequent deactivation of AP-1/TGF-β1 signaling pathway in neonatal rat cardiomyocytes. Thus, suppression of miR-21 prevents hypertrophic stimulation-induced cardiac remodeling by regulating PDCD4, AP-1, and TGF-β1 signaling pathway.
Collapse
|